US9198921B2 - Non-hormonal steroid modulators of NF-κB for treatment of disease - Google Patents

Non-hormonal steroid modulators of NF-κB for treatment of disease Download PDF

Info

Publication number
US9198921B2
US9198921B2 US13/639,650 US201113639650A US9198921B2 US 9198921 B2 US9198921 B2 US 9198921B2 US 201113639650 A US201113639650 A US 201113639650A US 9198921 B2 US9198921 B2 US 9198921B2
Authority
US
United States
Prior art keywords
disease
recited
group
muscular dystrophy
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US13/639,650
Other versions
US20130196962A1 (en
Inventor
John M. McCall
Eric Hoffman
Kanneboyina Nagaraju
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
FREERUN TECHNOLOGIES Inc
Reveragen Biopharma Inc
Original Assignee
Reveragen Biopharma Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Reveragen Biopharma Inc filed Critical Reveragen Biopharma Inc
Priority to US13/639,650 priority Critical patent/US9198921B2/en
Assigned to REVERAGEN BIOPHARMA, INC. reassignment REVERAGEN BIOPHARMA, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MCCALL, JOHN M, HOFFMAN, ERIC, NAGARAJU, KANNEBOYINA
Assigned to FREERUN TECHNOLOGIES, INC. reassignment FREERUN TECHNOLOGIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FORBES, FRANK, FRANZ, BENJAMIN JARED
Publication of US20130196962A1 publication Critical patent/US20130196962A1/en
Assigned to REVERAGEN BIOPHARMA, INC reassignment REVERAGEN BIOPHARMA, INC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: VALIDUS BIOPHARMA, INC.
Application granted granted Critical
Publication of US9198921B2 publication Critical patent/US9198921B2/en
Assigned to UNITED STATES GOVERNMENT reassignment UNITED STATES GOVERNMENT CONFIRMATORY LICENSE (SEE DOCUMENT FOR DETAILS). Assignors: REVERAGEN BIOPHARMA, INC.
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J9/00Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane
    • C07J9/005Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane containing a carboxylic function directly attached or attached by a chain containing only carbon atoms to the cyclopenta[a]hydrophenanthrene skeleton
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • A61K31/573Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone substituted in position 21, e.g. cortisone, dexamethasone, prednisone or aldosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/575Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of three or more carbon atoms, e.g. cholane, cholestane, ergosterol, sitosterol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J5/00Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond
    • C07J5/0007Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond not substituted in position 17 alfa
    • C07J5/0023Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond not substituted in position 17 alfa substituted in position 16
    • C07J5/003Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond not substituted in position 17 alfa substituted in position 16 by a saturated or unsaturated hydrocarbon group including 16-alkylidene substitutes
    • C07J5/0038Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond not substituted in position 17 alfa substituted in position 16 by a saturated or unsaturated hydrocarbon group including 16-alkylidene substitutes by an alkyl group
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J5/00Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond
    • C07J5/0046Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond substituted in position 17 alfa
    • C07J5/0053Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond substituted in position 17 alfa not substituted in position 16
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J5/00Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond
    • C07J5/0046Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond substituted in position 17 alfa
    • C07J5/0061Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond substituted in position 17 alfa substituted in position 16
    • C07J5/0069Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond substituted in position 17 alfa substituted in position 16 by a saturated or unsaturated hydrocarbon group
    • C07J5/0076Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond substituted in position 17 alfa substituted in position 16 by a saturated or unsaturated hydrocarbon group by an alkyl group
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J7/00Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of two carbon atoms
    • C07J7/008Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of two carbon atoms substituted in position 21
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J9/00Normal steroids containing carbon, hydrogen, halogen or oxygen substituted in position 17 beta by a chain of more than two carbon atoms, e.g. cholane, cholestane, coprostane
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J13/00Normal steroids containing carbon, hydrogen, halogen or oxygen having a carbon-to-carbon double bond from or to position 17
    • C07J13/005Normal steroids containing carbon, hydrogen, halogen or oxygen having a carbon-to-carbon double bond from or to position 17 with double bond in position 16 (17)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J21/00Normal steroids containing carbon, hydrogen, halogen or oxygen having an oxygen-containing hetero ring spiro-condensed with the cyclopenta(a)hydrophenanthrene skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J5/00Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond
    • C07J5/0007Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond not substituted in position 17 alfa
    • C07J5/0023Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond not substituted in position 17 alfa substituted in position 16
    • C07J5/003Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond not substituted in position 17 alfa substituted in position 16 by a saturated or unsaturated hydrocarbon group including 16-alkylidene substitutes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J5/00Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond
    • C07J5/0046Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond substituted in position 17 alfa
    • C07J5/0061Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond substituted in position 17 alfa substituted in position 16
    • C07J5/0069Normal steroids containing carbon, hydrogen, halogen or oxygen, substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane and substituted in position 21 by only one singly bound oxygen atom, i.e. only one oxygen bound to position 21 by a single bond substituted in position 17 alfa substituted in position 16 by a saturated or unsaturated hydrocarbon group
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07JSTEROIDS
    • C07J51/00Normal steroids with unmodified cyclopenta(a)hydrophenanthrene skeleton not provided for in groups C07J1/00 - C07J43/00
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • NF- ⁇ B non-hormonal steroid compounds and compositions and their application as pharmaceuticals for the treatment of disease.
  • Methods of modulation of NF- ⁇ B activity in a human or animal subject are also provided for the treatment of diseases mediated by NF- ⁇ B.
  • Muscular wasting diseases such as muscular dystrophies
  • muscular dystrophies are a group of degenerative diseases that culminate in progressive skeletal muscle wasting leading to muscle weakness, a high incidence of bone fracture, wheelchair dependence, and in some cases death.
  • Duchenne muscular dystrophy is the most severe and most widely recognized.
  • Another muscular wasting disease which shows similar symptoms, although less severe than Duchenne muscular dystrophy, is Becker muscular dystrophy. Even though the defective dystrophin gene causing both Duchenne muscular dystrophy and Becker muscular dystrophy has been known for over 20 years, a cure is still lacking.
  • TNF- ⁇ tumor necrosis factor-alpha
  • Muscle atrophy can also be induced by the loss of innervation or damage to innervation of the muscle tissue. Diseases such as chronic neuropathy and motor neuron disease can cause damage to innervation. Physical injury to the nerve can also lead to damage to the innervation of the muscle tissue.
  • muscle atrophy can be the result of environmental conditions such as during spaceflight or as a result of aging or extended bed rest. Under these environmental conditions, the muscles do not bear the usual weight load, resulting in muscle atrophy from disuse. Specifically, during muscle disuse, intracellular processes are activated to induce proteolysis, mainly through the ATP dependent ubiquitin proteasome pathway, which regulates the NF- ⁇ B pathway.
  • NF- ⁇ B is known to mediate extracellular signals responsible for the induction of genes involved in pro-inflammatory responses.
  • NF- ⁇ B is sequestered in the cytoplasm of most non-stimulated cells through a non-covalent interaction with one of several proteins known as inhibitors of kappa-beta (I ⁇ Bs) (May & Ghosh, (1997) Semin Cancer. Biol. 8, 63-73; May & Ghosh, (1998) Immunol. Today 19, 80-88; Ghosh et al., (1998) Annu. Rev. Immunol. 16, 225-260).
  • Cellular stimuli associated with pro-inflammatory responses such as TNF- ⁇ activate kinases, which in turn activate NF- ⁇ B by phosphorylating I ⁇ Bs.
  • IKKs I ⁇ B kinases
  • IKK ⁇ was initially identified as an I ⁇ B kinase induced by TNF- ⁇ stimulation in HeLa cells (DiDonato et al., (1997) Nature 388, 548-554).
  • IKK ⁇ Another I ⁇ B kinase homologous to IKK ⁇ was identified, termed IKK ⁇ , and determined to be the major I ⁇ B kinase induced following TNF ⁇ stimulation (Takeda et al., (1999) Science 284, 313-316; U.S. Pat. No.
  • IKK ⁇ and IKK ⁇ have an overall homology of 52% and a 65% homology in the kinase domain (Zandi et al., (1997) Cell 91, 243-252).
  • I ⁇ B protein kinases phosphorylate I ⁇ Bs at specific serine residues. Specifically, they phosphorylate serines 32 and 36 of I ⁇ B ⁇ (Traenckner et al., (1995) EMBO J. 14, 2876-2883; DiDonato et al., (1996) Mol. Cell. Biol. 16, 1295-1304). Phosphorylation of both sites is required to efficiently target I ⁇ B ⁇ for degradation.
  • IKK ⁇ and I ⁇ K ⁇ are usually in response to NF- ⁇ B activating agents including phorbol 12-myristate 13-acetate (PMA), lipopolysaccharide (LPS), interleukin-1 (IL-1), TNF- ⁇ , reactive oxygen species, and DNA damaging agents.
  • PMA phorbol 12-myristate 13-acetate
  • LPS lipopolysaccharide
  • IL-1 interleukin-1
  • IKK ⁇ and IKK ⁇ have distinct structural motifs including an amino terminal serine-threonine kinase domain separated from a carboxyl proximal helix-loop-helix domain by a leucine zipper domain. These structural characteristics are unlike other kinases, and the non-catalytic domains are thought to be involved in protein-protein interactions. As such, proteins which bind to IKKs should be capable of regulating the activity of NF- ⁇ B and potentially regulating downstream events such as induction of NF- ⁇ B.
  • NEMO NF- ⁇ B Essential Modulator
  • muscular wasting diseases may be treated in accordance with the present disclosure with a direct or indirect modulator of NF- ⁇ B.
  • Indirect modulators of NF- ⁇ B include, for example, inhibitors of I ⁇ B kinases (IKKs) such as IKK ⁇ inhibitors and IKK ⁇ inhibitors, and inhibitors functioning directly upstream from IKKs in the signaling pathway such as inhibitors of phosphoinositide dependent kinase (PDK) and inhibitors of Akt (also referred to as PKB).
  • IKKs I ⁇ B kinases
  • PDK phosphoinositide dependent kinase
  • Akt also referred to as PKB
  • IKKs I ⁇ B protein kinases
  • direct inhibiting compounds of IKK catalytic activity can be administered for the purpose of blocking the NF- ⁇ B pathway and inhibiting a muscular wasting disease.
  • inhibitors of IKK ⁇ or their enantiomers, analogs, prodrugs, active metabolites, salts, and/or hydrates thereof can be administered to the subject for the purpose of inhibiting a muscular wasting disease.
  • Novel compounds and pharmaceutical compositions certain of which have been found to modulate NF- ⁇ B have been discovered, together with methods of synthesizing and using the compounds including methods for the treatment of NF- ⁇ B-mediated diseases in a patient by administering the compounds.
  • said dashed line indicates an optional double bond
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of hydrogen, unsubstituted lower alkyl, lower haloalkyl, and halogen;
  • R 5 is selected from the group consisting of hydrogen, lower alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl, said lower alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkynyl, amido, amino, aryl, aryloxy, cycloalkyl, haloalkoxy, haloalkyl, heteroalkyl, heteroaryl, hydroxy, perhaloalkoxy, and thiol;
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and lower alkyl, said lower alkyl being optionally substituted with one or more substituents selected from the group consisting of alkenyl, alkoxy, alkyl, alkynyl, aryl, aryloxy, haloalkoxy, haloalkyl, heteroalkyl, hydroxy, and thiol;
  • R 7 and R 8 are independently selected from the groups consisting of hydrogen, unsubstituted C 1-3 alkyl, or R 7 and R 8 can be taken together to form oxo or C 3-6 saturated cycloalkyl;
  • R 9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkylamino, alkylthio, alkynyl, amido, amino, aryl, aryloxy, aroyl, carbamate, carboxyl, cyano, cycloalkyl, halogen, haloalkoxy, haloalkyl, heteroalkyl, heterocycloalkyl, heteroaryl, hydrazinyl, hydroxy, mercaptyl, nitro, oxo, perhaloalkoxy, sulfonate, alkylsulfonyl, N-sulfonamido, S-sulfonamido, and thiol.
  • substituents selected from the group consisting of acyl, alkenyl, alkoxy
  • R 7 and R 8 are independently selected from the groups consisting of hydrogen, unsubstituted C 2 -C 3 alkyl, or R 7 and R 8 can be taken together to form C 3-6 saturated cycloalkyl;
  • R 9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkylamino, alkylthio, alkynyl, amino, aryl, aryloxy, aroyl, carbamate, cyano, cycloalkyl, halogen, haloalkoxy, haloalkyl, heteroalkyl, heterocycloalkyl, heteroaryl, hydrazinyl, hydroxy, mercaptyl, perhaloalkoxy, sulfonate, alkylsulfonyl, N-sulfon
  • R 5 is selected from the group consisting of C 2 -C 8 alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl, said C 2 -C 8 alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkynyl, amido, amino, aryl, aryloxy, cycloalkyl, haloalkoxy, haloalkyl, heteroalkyl, heteroaryl, hydroxy, perhaloalkoxy, and thiol.
  • R 1 and R 3 are each hydrogen;
  • R 2 and R 4 are each independently selected from the group consisting of hydrogen, methyl, and fluorine;
  • R 5 is selected from the group consisting of hydrogen, unsubstituted lower alkyl, and phenyl;
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl;
  • R 7 and R 8 are each hydrogen;
  • R 9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino, hydroxyl, and carboxyl.
  • R 2 and R 4 are each hydrogen;
  • R 5 is selected from the group consisting of hydrogen, unsubstituted lower alkyl, and phenyl;
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl;
  • R 7 and R 8 are each hydrogen;
  • R 9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino, hydroxyl, and carboxyl.
  • R 5 is selected from the group consisting of hydrogen, methyl, and ethyl
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl
  • R 9 is selected from the group consisting of hydrogen, acetyl, and —C(O)CH 2 CH 2 CO 2 H.
  • R 5 is selected from the group consisting of unsubstituted C 2 -C 6 alkyl and phenyl;
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; and
  • R 9 is selected from the group consisting of hydrogen, acetyl, and —C(O)CH 2 CH 2 CO 2 H.
  • R 5 is ethyl
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of hydrogen, unsubstituted lower alkyl, lower haloalkyl, and halogen;
  • R 5 is selected from the group consisting of hydrogen, lower alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl, said lower alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkynyl, amido, amino, aryl, aryloxy, cycloalkyl, haloalkoxy, haloalkyl, heteroalkyl, heteroaryl, hydroxy, perhaloalkoxy, and thiol;
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and lower
  • R 7 and R 8 are independently selected from the groups consisting of hydrogen, unsubstituted C 2 -C 3 alkyl, or R 7 and R 8 can be taken together to form C 3-6 saturated cycloalkyl;
  • R 9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkylamino, alkylthio, alkynyl, amino, aryl, aryloxy, aroyl, carbamate, cyano, cycloalkyl, halogen, haloalkoxy, haloalkyl, heteroalkyl, heterocycloalkyl, heteroaryl, hydrazinyl, hydroxy, mercaptyl, perhaloalkoxy, alkylsulfonyl, N-sulfonamido, S-s
  • R 5 is selected from the group consisting of C 2 -C 8 alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl, said C 2 -C 8 alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkynyl, amido, amino, aryl, aryloxy, cycloalkyl, haloalkoxy, haloalkyl, heteroalkyl, heteroaryl, hydroxy, perhaloalkoxy, and thiol.
  • R 1 and R 3 are each hydrogen;
  • R 2 and R 4 are each independently selected from the group consisting of hydrogen, methyl, and fluorine;
  • R 5 is selected from the group consisting of hydrogen, unsubstituted lower alkyl, and phenyl;
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl;
  • R 7 and R 8 are each hydrogen;
  • R 9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino, hydroxyl, and carboxyl.
  • R 2 and R 4 are each hydrogen;
  • R 5 is selected from the group consisting of hydrogen, unsubstituted lower alkyl, and phenyl;
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl;
  • R 7 and R 8 are each hydrogen;
  • R 9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino, hydroxyl, and carboxyl.
  • R 5 is selected from the group consisting of hydrogen, methyl, and ethyl
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl
  • R 9 is selected from the group consisting of hydrogen, acetyl, and —C(O)CH 2 CH 2 CO 2 H.
  • R 5 is selected from the group consisting of unsubstituted C 2 -C 6 alkyl and phenyl;
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; and
  • R 9 is selected from the group consisting of hydrogen, acetyl, and —C(O)CH 2 CH 2 CO 2 H.
  • R 5 is ethyl
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of hydrogen, unsubstituted lower alkyl, lower haloalkyl, and halogen;
  • R 5 is selected from the group consisting of hydrogen, lower alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl, said lower alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkynyl, amido, amino, aryl, aryloxy, cycloalkyl, haloalkoxy, haloalkyl, heteroalkyl, heteroaryl, hydroxy, perhaloalkoxy, and thiol;
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and lower
  • R 1 and R 3 are each hydrogen;
  • R 2 and R 4 are each independently selected from the group consisting of hydrogen, methyl, and fluorine;
  • R 5 is selected from the group consisting of hydrogen, unsubstituted lower alkyl, and phenyl;
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl;
  • R 7 and R 8 are each hydrogen;
  • R 9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino and hydroxyl.
  • R 2 and R 4 are each hydrogen;
  • R 5 is selected from the group consisting of hydrogen, unsubstituted lower alkyl, and phenyl;
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl;
  • R 7 and R 8 are each hydrogen; and
  • R 9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino and hydroxyl.
  • R 5 is selected from the group consisting of hydrogen, methyl, and ethyl
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl
  • R 9 is selected from the group consisting of hydrogen and acetyl.
  • R 1 , R 2 , R 3 , and R 4 are each independently selected from the group consisting of hydrogen, unsubstituted lower alkyl, lower haloalkyl, and halogen;
  • R 5 is selected from the group consisting of C 2 -C 8 alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl, said C 2 -C 8 alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkynyl, amido, amino, aryl, aryloxy, cycloalkyl, haloalkoxy, haloalkyl, heteroalkyl, heteroaryl, hydroxy, perhaloalkoxy, and thiol;
  • R 6 is selected from the group consisting of
  • R 1 and R 3 are each hydrogen;
  • R 2 and R 4 are each independently selected from the group consisting of hydrogen, methyl, and fluorine;
  • R 5 is selected from the group consisting of unsubstituted C 2 -C 6 alkyl and phenyl;
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl;
  • R 7 and R 8 are each hydrogen;
  • R 9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino, hydroxyl, and carboxyl.
  • R 2 and R 4 are each hydrogen; R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; R 7 and R 8 are each hydrogen; and R 9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino, hydroxyl, and carboxyl.
  • R 6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; and R 9 is selected from the group consisting of hydrogen, acetyl, and —C(O)CH 2 CH 2 CO 2 H.
  • R 5 is ethyl
  • Certain compounds disclosed herein may possess useful NF- ⁇ B modulating activity, and may be used in the treatment or prophylaxis of a disease or condition in which NF- ⁇ B plays an active role.
  • certain embodiments also provide pharmaceutical compositions comprising one or more compounds disclosed herein together with a pharmaceutically acceptable carrier, as well as methods of making and using the compounds and compositions.
  • Certain embodiments provide methods for modulating NF- ⁇ B.
  • Other embodiments provide methods for treating a NF- ⁇ B-mediated disorder in a patient in need of such treatment, comprising administering to said patient a therapeutically effective amount of a compound or composition according to the present invention.
  • certain compounds disclosed herein for use in the manufacture of a medicament for the treatment of a disease or condition ameliorated by the modulation of NF- ⁇ B.
  • said NF- ⁇ B-mediated disease is selected from the group consisting of muscular dystrophy, arthritis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type 2 diabetes, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, muscle wasting, catabolic disorders, obesity, fetal growth retardation, hypercholesterolemia, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, asthma, chronic obstructive pulmonary disease, Sjogren's syndrome, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, gut diseases, peritoneal endometriosis, skin diseases, nasal sinusitis, mesothelioma, anhidrotic ecodermal dysp
  • said disease is said NF- ⁇ B-mediated disease is asthma or chronic obstructive pulmonary disease.
  • said NF- ⁇ B-mediated is Sjogren's syndrome.
  • said NF- ⁇ B-mediated is arthritis.
  • said NF- ⁇ B-mediated is muscular wasting.
  • said muscular wasting disease is muscular dystrophy.
  • said muscular dystrophy is selected from the group consisting of Duchenne muscular dystrophy, Becker muscular dystrophy, limb girdle muscular dystrophy, congenital muscular dystrophy, facioscapulohumeral muscular dystrophy, myotonic muscular dystrophy, oculopharyngeal muscular dystrophy, distal muscular dystrophy, and Emery-Dreifuss muscular dystrophy.
  • said muscular dystrophy is Duchenne muscular dystrophy.
  • disclosed herein is a compound selected from the group consisting of examples 25-40.
  • a pharmaceutical composition comprising a compound selected from the group consisting of examples 25-40 together with a pharmaceutically acceptable carrier.
  • said pharmaceutical composition comprises a tablet or a capsule.
  • said pharmaceutical composition comprises another therapeutic agent.
  • a disease selected from the group consisting of muscular dystrophy, arthritis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type 2 diabetes, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, muscle wasting, catabolic disorders, obesity, fetal growth retardation, hypercholesterolemia, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, asthma, chronic obstructive pulmonary disease, Sjogren's syndrome, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, gut diseases, peritoneal endometriosis, skin diseases, nasal sinusitis, mesothelioma, anhidr
  • a disease selected from the group consisting of muscular dystrophy, arthritis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type 2 diabetes, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, muscle wasting, catabolic disorders, obesity, fetal growth retardation, hypercholesterolemia, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, asthma, chronic obstructive pulmonary disease, Sjogren's syndrome, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, gut diseases, peritoneal endometriosis, skin diseases, nasal sinusitis, mesothelioma, anhidr
  • said disease is muscular dystrophy.
  • said muscular dystrophy is selected from the group consisting of Duchenne muscular dystrophy, Becker muscular dystrophy, limb girdle muscular dystrophy, congenital muscular dystrophy, facioscapulohumeral muscular dystrophy, myotonic muscular dystrophy, oculopharyngeal muscular dystrophy, distal muscular dystrophy, and Emery-Dreifuss muscular dystrophy.
  • said muscular dystrophy is Duchenne muscular dystrophy.
  • said disease is selected from the group consisting of asthma and chronic obstructive pulmonary disease.
  • said disease is asthma.
  • said disease is chronic obstructive pulmonary disease.
  • said disease is arthritis.
  • said disease is selected from the group consisting of traumatic brain injury and spinal cord injury.
  • said disease is traumatic brain injury.
  • said disease is spinal cord injury.
  • said disease is amyotropic lateral sclerosis.
  • said disease is Sjogren's syndrome.
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said compound has the structural formula:
  • said method comprises the administration of another therapeutic agent.
  • a compound selected from the group consisting of examples 25-40 for use as a medicament is a compound selected from the group consisting of examples 25-40 for use as a medicament.
  • a disease selected from the group
  • acyl refers to a carbonyl attached to an alkenyl, alkyl, aryl, cycloalkyl, heteroaryl, heterocycle, or any other moiety were the atom attached to the carbonyl is carbon.
  • An “acetyl” group refers to a —C(O)CH 3 group.
  • An “alkylcarbonyl” or “alkanoyl” group refers to an alkyl group attached to the parent molecular moiety through a carbonyl group. Examples of such groups include methylcarbonyl and ethylcarbonyl. Examples of acyl groups include formyl, alkanoyl and aroyl.
  • alkenyl refers to a straight-chain or branched-chain hydrocarbon group having one or more double bonds and containing from 2 to 20 carbon atoms. In certain embodiments, said alkenyl will comprise from 2 to 6 carbon atoms.
  • alkenylene refers to a carbon-carbon double bond system attached at two or more positions such as ethenylene [(—CH ⁇ CH—),(—C::C—)]. Examples of suitable alkenyl groups include ethenyl, propenyl, 2-methylpropenyl, 1,4-butadienyl and the like. Unless otherwise specified, the term “alkenyl” may include “alkenylene” groups.
  • alkoxy refers to an alkyl ether group, wherein the term alkyl is as defined below.
  • suitable alkyl ether groups include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, iso-butoxy, sec-butoxy, tert-butoxy, and the like.
  • alkyl refers to a straight-chain or branched-chain alkyl group containing from 1 to 20 carbon atoms. In certain embodiments, said alkyl will comprise from 1 to 10 carbon atoms. In further embodiments, said alkyl will comprise from 1 to 6 carbon atoms. Alkyl groups may be optionally substituted as defined herein. Examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, iso-amyl, hexyl, octyl, noyl and the like.
  • alkylene refers to a saturated aliphatic group derived from a straight or branched chain saturated hydrocarbon attached at two or more positions, such as methylene (—CH 2 —). Unless otherwise specified, the term “alkyl” may include “alkylene” groups.
  • alkylamino refers to an alkyl group attached to the parent molecular moiety through an amino group. Suitable alkylamino groups may be mono- or dialkylated, forming groups such as, for example, N-methylamino, N-ethylamino, N,N-dimethylamino, N,N-ethylmethylamino and the like.
  • alkylidene refers to an alkenyl group in which one carbon atom of the carbon-carbon double bond belongs to the moiety to which the alkenyl group is attached.
  • alkylthio refers to an alkyl thioether (R—S—) group wherein the term alkyl is as defined above and wherein the sulfur may be singly or doubly oxidized.
  • suitable alkyl thioether groups include methylthio, ethylthio, n-propylthio, isopropylthio, n-butylthio, iso-butylthio, sec-butylthio, tert-butylthio, methanesulfonyl, ethanesulfinyl, and the like.
  • alkynyl refers to a straight-chain or branched chain hydrocarbon group having one or more triple bonds and containing from 2 to 20 carbon atoms. In certain embodiments, said alkynyl comprises from 2 to 6 carbon atoms. In further embodiments, said alkynyl comprises from 2 to 4 carbon atoms.
  • alkynylene refers to a carbon-carbon triple bond attached at two positions such as ethynylene (—C:::C—, —C ⁇ C—).
  • alkynyl groups include ethynyl, propynyl, hydroxypropynyl, butyn-1-yl, butyn-2-yl, pentyn-1-yl, 3-methylbutyn-1-yl, hexyn-2-yl, and the like.
  • alkynyl may include “alkynylene” groups.
  • acylamino as used herein, alone or in combination, embraces an acyl group attached to the parent moiety through an amino group.
  • An example of an “acylamino” group is acetylamino (CH 3 C(O)NH—).
  • amino refers to —NRR′, wherein R and R′ are independently selected from the group consisting of hydrogen, alkyl, acyl, heteroalkyl, aryl, cycloalkyl, heteroaryl, and heterocycloalkyl, any of which may themselves be optionally substituted. Additionally, R and R′ may combine to form heterocycloalkyl, either of which may be optionally substituted.
  • aryl as used herein, alone or in combination, means a carbocyclic aromatic system containing one, two or three rings wherein such polycyclic ring systems are fused together.
  • aryl embraces aromatic groups such as phenyl, naphthyl, anthracenyl, and phenanthryl.
  • arylalkenyl or “aralkenyl,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkenyl group.
  • arylalkoxy or “aralkoxy,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkoxy group.
  • arylalkyl or “aralkyl,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkyl group.
  • arylalkynyl or “aralkynyl,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkynyl group.
  • arylalkanoyl or “aralkanoyl” or “aroyl,” as used herein, alone or in combination, refers to an acyl group derived from an aryl-substituted alkanecarboxylic acid such as benzoyl, napthoyl, phenylacetyl, 3-phenylpropionyl (hydrocinnamoyl), 4-phenylbutyryl, (2-naphthyl)acetyl, 4-chlorohydrocinnamoyl, and the like.
  • an aryl-substituted alkanecarboxylic acid such as benzoyl, napthoyl, phenylacetyl, 3-phenylpropionyl (hydrocinnamoyl), 4-phenylbutyryl, (2-naphthyl)acetyl, 4-chlorohydrocinnamoyl, and the like.
  • aryloxy refers to an aryl group attached to the parent molecular moiety through an oxy.
  • carbamate refers to an ester of carbamic acid (—NHCOO—) which may be attached to the parent molecular moiety from either the nitrogen or acid end, and which may be optionally substituted as defined herein.
  • O-carbamyl as used herein, alone or in combination, refers to a —OC(O)NRR′, group-with R and R′ as defined herein.
  • N-carbamyl as used herein, alone or in combination, refers to a ROC(O)NR′— group, with R and R′ as defined herein.
  • carbonyl when alone includes formyl [—C(O)H] and in combination is a —C(O)— group.
  • carboxyl or “carboxy,” as used herein, refers to —C(O)OH or the corresponding “carboxylate” anion, such as is in a carboxylic acid salt.
  • An “O-carboxy” group refers to a RC(O)O— group, where R is as defined herein.
  • a “C-carboxy” group refers to a —C(O)OR groups where R is as defined herein.
  • cyano as used herein, alone or in combination, refers to —CN.
  • cycloalkyl groups examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, tetrahydronapthyl, indanyl, octahydronaphthyl, 2,3-dihydro-1H-indenyl, adamantyl and the like.
  • “Bicyclic” and “tricyclic” as used herein are intended to include both fused ring systems, such as decahydronaphthalene, octahydronaphthalene as well as the multicyclic (multicentered) saturated or partially unsaturated type. The latter type of isomer is exemplified in general by, bicyclo[1,1,1]pentane, camphor, adamantane, and bicyclo[3,2,1]octane.
  • esters refers to a carboxy group bridging two moieties linked at carbon atoms.
  • ether refers to an oxy group bridging two moieties linked at carbon atoms.
  • halo or halogen, as used herein, alone or in combination, refers to fluorine, chlorine, bromine, or iodine.
  • haloalkoxy refers to a haloalkyl group attached to the parent molecular moiety through an oxygen atom.
  • haloalkyl groups include fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl, difluoropropyl, dichloroethyl and dichloropropyl.
  • Haloalkylene refers to a haloalkyl group attached at two or more positions. Examples include fluoromethylene (—CFH—), difluoromethylene (—CF 2 —), chloromethylene (—CHCl—) and the like.
  • heteroalkyl refers to a stable straight or branched chain, or cyclic hydrocarbon group, or combinations thereof, fully saturated or containing from 1 to 3 degrees of unsaturation, consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of O, N, and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N and S may be placed at any interior position of the heteroalkyl group. Up to two heteroatoms may be consecutive, such as, for example, —CH 2 —NH—OCH 3 .
  • heteroaryl refers to a 3 to 7 membered unsaturated heteromonocyclic ring, or a fused monocyclic, bicyclic, or tricyclic ring system in which at least one of the fused rings is aromatic, which contains at least one atom selected from the group consisting of O, S, and N.
  • said heteroaryl will comprise from 5 to 7 carbon atoms.
  • heterocyclic rings are fused with aryl rings, wherein heteroaryl rings are fused with other heteroaryl rings, wherein heteroaryl rings are fused with heterocycloalkyl rings, or wherein heteroaryl rings are fused with cycloalkyl rings.
  • heteroaryl groups include pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, pyranyl, furyl, thienyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, thiadiazolyl, isothiazolyl, indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, quinoxalinyl, quinazolinyl, indazolyl, benzotriazolyl, benzodioxolyl, benzopyranyl, benzoxazolyl, benzoxadiazolyl, benzothiazolyl, benzothiadiazolyl, benzofuryl, benzothienyl, chromonyl,
  • Exemplary tricyclic heterocyclic groups include carbazolyl, benzindolyl, phenanthrolinyl, dibenzofuranyl, acridinyl, phenanthridinyl, xanthenyl and the like.
  • heterocycloalkyl and, interchangeably, “heterocycle,” as used herein, alone or in combination, each refer to a saturated, partially unsaturated, or fully unsaturated monocyclic, bicyclic, or tricyclic heterocyclic group containing at least one heteroatom as a ring member, wherein each said heteroatom may be independently selected from the group consisting of nitrogen, oxygen, and sulfur
  • said heterocycloalkyl will comprise from 1 to 4 heteroatoms as ring members.
  • said heterocycloalkyl will comprise from 1 to 2 heteroatoms as ring members.
  • said heterocycloalkyl will comprise from 3 to 8 ring members in each ring.
  • heterocycloalkyl will comprise from 3 to 7 ring members in each ring. In yet further embodiments, said heterocycloalkyl will comprise from 5 to 6 ring members in each ring.
  • “Heterocycloalkyl” and “heterocycle” are intended to include sulfones, sulfoxides, N-oxides of tertiary nitrogen ring members, and carbocyclic fused and benzo fused ring systems; additionally, both terms also include systems where a heterocycle ring is fused to an aryl group, as defined herein, or an additional heterocycle group.
  • heterocycle groups include aziridinyl, azetidinyl, 1,3-benzodioxolyl, dihydroisoindolyl, dihydroisoquinolinyl, dihydrocinnolinyl, dihydrobenzodioxinyl, dihydro[1,3]oxazolo[4,5-b]pyridinyl, benzothiazolyl, dihydroindolyl, dihy-dropyridinyl, 1,3-dioxanyl, 1,4-dioxanyl, 1,3-dioxolanyl, isoindolinyl, morpholinyl, piperazinyl, pyrrolidinyl, tetrahydropyridinyl, piperidinyl, thiomorpholinyl, and the like.
  • the heterocycle groups may be optionally substituted unless specifically prohibited.
  • hydrazinyl as used herein, alone or in combination, refers to two amino groups joined by a single bond, i.e., —N—N—.
  • hydroxyalkyl refers to a hydroxy group attached to the parent molecular moiety through an alkyl group.
  • amino as used herein, alone or in combination, refers to ⁇ N—.
  • aminohydroxy refers to ⁇ N(OH) and ⁇ N—O—.
  • the phrase “in the main chain” refers to the longest contiguous or adjacent chain of carbon atoms starting at the point of attachment of a group to the compounds of any one of the formulas disclosed herein.
  • isocyanato refers to a —NCO group.
  • isothiocyanato refers to a —NCS group.
  • linear chain of atoms refers to the longest straight chain of atoms independently selected from carbon, nitrogen, oxygen and sulfur.
  • lower means containing from 1 to and including 6 carbon atoms.
  • lower aryl as used herein, alone or in combination, means phenyl or naphthyl, which may be optionally substituted as provided.
  • lower heteroaryl means either 1) monocyclic heteroaryl comprising five or six ring members, of which between one and four said members may be heteroatoms selected from the group consisting of O, S, and N, or 2) bicyclic heteroaryl, wherein each of the fused rings comprises five or six ring members, comprising between them one to four heteroatoms selected from the group consisting of O, S, and N.
  • lower cycloalkyl means a monocyclic cycloalkyl having between three and six ring members. Lower cycloalkyls may be unsaturated. Examples of lower cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • lower heterocycloalkyl means a monocyclic heterocycloalkyl having between three and six ring members, of which between one and four may be heteroatoms selected from the group consisting of O, S, and N.
  • lower heterocycloalkyls include pyrrolidinyl, imidazolidinyl, pyrazolidinyl, piperidinyl, piperazinyl, and morpholinyl.
  • Lower heterocycloalkyls may be unsaturated.
  • lower amino refers to —NRR′, wherein R and R′ are independently selected from the group consisting of hydrogen, lower alkyl, and lower heteroalkyl, any of which may be optionally substituted. Additionally, the R and R′ of a lower amino group may combine to form a five- or six-membered heterocycloalkyl, either of which may be optionally substituted.
  • mercaptyl as used herein, alone or in combination, refers to an RS— group, where R is as defined herein.
  • nitro refers to —NO 2 .
  • oxy or “oxa,” as used herein, alone or in combination, refer to —O—.
  • perhaloalkoxy refers to an alkoxy group where all of the hydrogen atoms are replaced by halogen atoms.
  • perhaloalkyl refers to an alkyl group where all of the hydrogen atoms are replaced by halogen atoms.
  • sulfonate refers the —SO 3 H group and its anion as the sulfonic acid is used in salt formation.
  • sulfonyl as used herein, alone or in combination, refers to —S(O) 2 —.
  • N-sulfonamido refers to a RS( ⁇ O) 2 NR′— group with R and R′ as defined herein.
  • S-sulfonamido refers to a —S( ⁇ O) 2 NRR′, group, with R and
  • thia and thio refer to a —S— group or an ether wherein the oxygen is replaced with sulfur.
  • the oxidized derivatives of the thio group namely sulfinyl and sulfonyl, are included in the definition of thia and thio.
  • thiol as used herein, alone or in combination, refers to an —SH group.
  • thiocarbonyl when alone includes thioformyl —C(S)H and in combination is a —C(S)— group.
  • N-thiocarbamyl refers to an ROC(S)NR′— group, with R and R′ as defined herein.
  • O-thiocarbamyl refers to a —OC(S)NRR′, group with R and R′ as defined herein.
  • thiocyanato refers to a —CNS group.
  • trihalomethanesulfonamido refers to a X 3 CS(O) 2 NR— group with X is a halogen and R as defined herein.
  • trihalomethanesulfonyl refers to a X 3 CS(O) 2 — group where X is a halogen.
  • trihalomethoxy refers to a X 3 CO— group where X is a halogen.
  • trimethysilyl as used herein, alone or in combination, refers to a silicone group substituted at its three free valences with groups as listed herein under the definition of substituted amino. Examples include trimethysilyl, tert-butyldimethylsilyl, triphenylsilyl and the like.
  • any definition herein may be used in combination with any other definition to describe a composite structural group.
  • the trailing element of any such definition is that which attaches to the parent moiety.
  • the composite group alkylamido would represent an alkyl group attached to the parent molecule through an amido group
  • the term alkoxyalkyl would represent an alkoxy group attached to the parent molecule through an alkyl group.
  • the term “optionally substituted” means the anteceding group may be substituted or unsubstituted.
  • the substituents of an “optionally substituted” group may include, without limitation, one or more substituents independently selected from the following groups or a particular designated set of groups, alone or in combination: lower alkyl, lower alkenyl, lower alkynyl, lower alkanoyl, lower heteroalkyl, lower heterocycloalkyl, lower haloalkyl, lower haloalkenyl, lower haloalkynyl, lower perhaloalkyl, lower perhaloalkoxy, lower cycloalkyl, phenyl, aryl, aryloxy, lower alkoxy, lower haloalkoxy, oxo, lower acyloxy, carbonyl, carboxyl, lower alkylcarbonyl, lower carboxyester, lower carboxamido, cyano, hydrogen, halogen, hydroxy, amino, lower alkylamino
  • Two substituents may be joined together to form a fused five-, six-, or seven-membered carbocyclic or heterocyclic ring consisting of zero to three heteroatoms, for example forming methylenedioxy or ethylenedioxy.
  • An optionally substituted group may be unsubstituted (e.g., —CH 2 CH 3 ), fully substituted (e.g., —CF 2 CF 3 ), monosubstituted (e.g., —CH 2 CH 2 F) or substituted at a level anywhere in-between fully substituted and monosubstituted (e.g., —CH 2 CF 3 ).
  • R or the term R′ refers to a moiety selected from the group consisting of hydrogen, alkyl, cycloalkyl, heteroalkyl, aryl, heteroaryl and heterocycloalkyl, any of which may be optionally substituted.
  • aryl, heterocycle, R, etc. occur more than one time in a formula or generic structure, its definition at each occurrence is independent of the definition at every other occurrence.
  • certain groups may be attached to a parent molecule or may occupy a position in a chain of elements from either end as written.
  • an unsymmetrical group such as —C(O)N(R)— may be attached to the parent moiety at either the carbon or the nitrogen.
  • bonds refers to a covalent linkage between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure.
  • a bond may be single, double, or triple unless otherwise specified.
  • a dashed line between two atoms in a drawing of a molecule indicates that an additional bond may be present or absent at that position.
  • disease as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disorder” and “condition” (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms, and causes the human or animal to have a reduced duration or quality of life.
  • NF- ⁇ B-mediated disease refers to a disease in which NF- ⁇ B plays an active role in the disease pathology.
  • NF- ⁇ B-mediated diseases include diseases in which multiple biological pathways and/or processes in addition to NF- ⁇ B-mediated processes contribute to the disease pathology.
  • a NF- ⁇ B-mediated disease may be completely or partially mediated by modulating the activity or amount of NF- ⁇ B.
  • a NF- ⁇ B-mediated disease is one in which modulation of NF- ⁇ B results in some effect on the underlying disease e.g., administration of a NF- ⁇ B modulator results in some improvement in at least some of the patients being treated.
  • NF- ⁇ B-mediated disease also refers to the following diseases, even though the compounds disclosed herein exert their effects through biological pathways and/or processes other than NF- ⁇ B: muscular dystrophy, arthritis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type 2 diabetes, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, muscle wasting, catabolic disorders, obesity, fetal growth retardation, hypercholesterolemia, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, asthma, chronic obstructive pulmonary disease, Sjogren's syndrome, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, gut diseases, peritoneal endometriosis, skin diseases,
  • combination therapy means the administration of two or more therapeutic agents to treat a therapeutic condition or disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.
  • NF- ⁇ B modulator is used herein to refer to a compound that exhibits an EC 50 with respect to NF- ⁇ B activity of no more than about 100 ⁇ M and more typically not more than about 50 ⁇ M, as measured in the NF- ⁇ B inhibitor assays described generally hereinbelow.
  • EC 50 is that concentration of modulator which either activates or reduces the activity or increases or decreases the amount of an enzyme (e.g., (NF- ⁇ B)) to half-maximal level. Certain compounds disclosed herein have been discovered to exhibit modulatory activity against NF- ⁇ B.
  • compounds will exhibit an EC 50 with respect to NF- ⁇ B of no more than about 10 ⁇ M; in further embodiments, compounds will exhibit an EC 50 with respect to NF- ⁇ B of no more than about 5 ⁇ M; in yet further embodiments, compounds will exhibit an EC 50 with respect to NF- ⁇ B of not more than about 1 ⁇ M; in yet further embodiments, compounds will exhibit an EC 50 with respect to NF- ⁇ B of not more than about 200 nM, as measured in the NF- ⁇ B assay described herein.
  • terapéuticaally effective is intended to qualify the amount of active ingredients used in the treatment of a disease or disorder. This amount will achieve the goal of reducing or eliminating the said disease or disorder.
  • terapéuticaally acceptable refers to those compounds (or salts, prodrugs, tautomers, zwitterionic forms, etc.) which are suitable for use in contact with the tissues of patients without undue toxicity, irritation, and allergic response, are commensurate with a reasonable benefit/risk ratio, and are effective for their intended use.
  • patient means all mammals including humans. Examples of patients include humans, cows, dogs, cats, goats, sheep, pigs, and rabbits. Preferably, the patient is a human.
  • prodrug refers to a compound that is made more active in vivo.
  • Certain compounds disclosed herein may also exist as prodrugs, as described in Hydrolysis in Drug and Prodrug Metabolism: Chemistry, Biochemistry, and Enzymology (Testa, Bernard and Mayer, Joachim M. Wiley-VHCA, Zurich, Switzerland 2003).
  • Prodrugs of the compounds described herein are structurally modified forms of the compound that readily undergo chemical changes under physiological conditions to provide the compound.
  • prodrugs can be converted to the compound by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to a compound when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.
  • Prodrugs are often useful because, in some situations, they may be easier to administer than the compound, or parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent drug.
  • a wide variety of prodrug derivatives are known in the art, such as those that rely on hydrolytic cleavage or oxidative activation of the prodrug.
  • An example, without limitation, of a prodrug would be a compound which is administered as an ester (the “prodrug”), but then is metabolically hydrolyzed to the carboxylic acid, the active entity. Additional examples include peptidyl derivatives of a compound.
  • the compounds disclosed herein can exist as therapeutically acceptable salts.
  • the present invention includes compounds listed above in the form of salts, including acid addition salts. Suitable salts include those formed with both organic and inorganic acids. Such acid addition salts will normally be pharmaceutically acceptable. However, salts of non-pharmaceutically acceptable salts may be of utility in the preparation and purification of the compound in question. Basic addition salts may also be formed and be pharmaceutically acceptable. For a more complete discussion of the preparation and selection of salts, refer to Pharmaceutical Salts Properties, Selection, and Use (Stahl, P. Heinrich. Wiley-VCHA, Zurich, Switzerland, 2002).
  • terapéuticaally acceptable salt represents salts or zwitterionic forms of the compounds disclosed herein which are water or oil-soluble or dispersible and therapeutically acceptable as defined herein.
  • the salts can be prepared during the final isolation and purification of the compounds or separately by reacting the appropriate compound in the form of the free base with a suitable acid.
  • Representative acid addition salts include acetate, adipate, alginate, L-ascorbate, aspartate, benzoate, benzenesulfonate (besylate), bisulfate, butyrate, camphorate, camphorsulfonate, citrate, digluconate, formate, fumarate, gentisate, glutarate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethansulfonate (isethionate), lactate, maleate, malonate, DL-mandelate, mesitylenesulfonate, methanesulfonate, naphthylenesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenyl
  • basic groups in the compounds disclosed herein can be quaternized with methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides; dimethyl, diethyl, dibutyl, and diamyl sulfates; decyl, lauryl, myristyl, and steryl chlorides, bromides, and iodides; and benzyl and phenethyl bromides.
  • acids which can be employed to form therapeutically acceptable addition salts include inorganic acids such as hydrochloric, hydrobromic, sulfuric, and phosphoric, and organic acids such as oxalic, maleic, succinic, and citric. Salts can also be formed by coordination of the compounds with an alkali metal or alkaline earth ion.
  • the present invention contemplates sodium, potassium, magnesium, and calcium salts of the compounds disclosed herein, and the like.
  • Basic addition salts can be prepared during the final isolation and purification of the compounds by reacting a carboxy group with a suitable base such as the hydroxide, carbonate, or bicarbonate of a metal cation or with ammonia or an organic primary, secondary, or tertiary amine.
  • a suitable base such as the hydroxide, carbonate, or bicarbonate of a metal cation or with ammonia or an organic primary, secondary, or tertiary amine.
  • the cations of therapeutically acceptable salts include lithium, sodium, potassium, calcium, magnesium, and aluminum, as well as nontoxic quaternary amine cations such as ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, diethylamine, ethylamine, tributylamine, pyridine, N,N-dimethylaniline, N-methylpiperidine, N-methylmorpholine, dicyclohexylamine, procaine, dibenzylamine, N,N-dibenzylphenethylamine, 1-ephenamine, and N,N′-dibenzylethylenediamine.
  • Other representative organic amines useful for the formation of base addition salts include ethylenediamine, ethanolamine, diethanolamine, piperidine, and piperazine.
  • the salts may include hydrochloride, hydrobromide, sulfonate, citrate, tartrate, phosphonate, lactate, pyruvate, acetate, succinate, oxalate, fumarate, malate, oxaloacetate, methanesulfonate, ethanesulfonate, p-toluenesulfonate, benzenesulfonate and isethionate salts of compounds disclosed herein.
  • a salt of a compound can be made by reacting the appropriate compound in the form of the free base with the appropriate acid.
  • compositions which comprise one or more of certain compounds disclosed herein, or one or more pharmaceutically acceptable salts, esters, prodrugs, amides, or solvates thereof, together with one or more pharmaceutically acceptable carriers thereof and optionally one or more other therapeutic ingredients.
  • the carrier(s) must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., in Remington's Pharmaceutical Sciences.
  • compositions disclosed herein may be manufactured in any manner known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
  • the formulations include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous, intraarticular, and intramedullary), intraperitoneal, transmucosal, transdermal, rectal and topical (including dermal, buccal, sublingual and intraocular) administration although the most suitable route may depend upon for example the condition and disorder of the recipient.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Typically, these methods include the step of bringing into association a compound of the subject invention or a pharmaceutically acceptable salt, ester, amide, prodrug or solvate thereof (“active ingredient”) with the carrier which constitutes one or more accessory ingredients.
  • active ingredient a pharmaceutically acceptable salt, ester, amide, prodrug or solvate thereof
  • the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
  • Formulations of the compounds disclosed herein suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • compositions which can be used orally include tablets, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. Tablets may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with binders, inert diluents, or lubricating, surface active or dispersing agents. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein. All formulations for oral administration should be in dosages suitable for such administration.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added.
  • Dragee cores are provided with suitable coatings.
  • concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • the compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in powder form or in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or sterile pyrogen-free water, immediately prior to use.
  • sterile liquid carrier for example, saline or sterile pyrogen-free water
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • Formulations for parenteral administration include aqueous and non-aqueous (oily) sterile injection solutions of the active compounds which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the compounds may be formulated as nanoparticle preparations.
  • Such nanoparticle preparations can include, for example, nanoshere encapsulations of active compounds, inactive nanoparticles to which active compounds can be tethered, or nanoscale powders of active compounds. Nanoparticle preparations can be used to increase the bioavailability of the active compounds, control the rate of release of the active compounds, or deliver active compounds to a particular location in the body. See A. Dove, “An Easy Pill to Swallow”, Drug Discovery & Development Magazine: 11(11), November, 2008, pp. 22-24.
  • the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • compositions may take the form of tablets, lozenges, pastilles, or gels formulated in conventional manner.
  • Such compositions may comprise the active ingredient in a flavored basis such as sucrose and acacia or tragacanth.
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter, polyethylene glycol, or other glycerides.
  • Certain compounds disclosed herein may be administered topically, that is by non-systemic administration. This includes the application of a compound disclosed herein externally to the epidermis or the buccal cavity and the instillation of such a compound into the ear, eye and nose, such that the compound does not significantly enter the blood stream.
  • systemic administration refers to oral, intravenous, intraperitoneal and intramuscular administration.
  • Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin to the site of inflammation such as gels, liniments, lotions, creams, ointments or pastes, and drops suitable for administration to the eye, ear or nose.
  • the active ingredient for topical administration may comprise, for example, from 0.001% to 10% w/w (by weight) of the formulation. In certain embodiments, the active ingredient may comprise as much as 10% w/w. In other embodiments, it may comprise less than 5% w/w. In certain embodiments, the active ingredient may comprise from 2% w/w to 5% w/w. In other embodiments, it may comprise from 0.1% to 1% w/w of the formulation.
  • Formulations for topical administration in the mouth include lozenges comprising the active ingredient in a flavored basis such as sucrose and acacia or tragacanth, and pastilles comprising the active ingredient in a basis such as gelatin and glycerin or sucrose and acacia.
  • compounds may be conveniently delivered from an insufflator, nebulizer pressurized packs or other convenient means of delivering an aerosol spray.
  • Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • the compounds according to the invention may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the powder composition may be presented in unit dosage form, in for example, capsules, cartridges, gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflator.
  • Preferred unit dosage formulations are those containing an effective dose, as herein below recited, or an appropriate fraction thereof, of the active ingredient.
  • formulations described above may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
  • Compounds may be administered orally or via injection at a dose of from 0.1 to 500 mg/kg per day.
  • the dose range for adult humans is generally from 5 mg to 2 g/day.
  • Tablets or other forms of presentation provided in discrete units may conveniently contain an amount of one or more compounds which is effective at such dosage or as a multiple of the same, for instance, units containing 5 mg to 500 mg, usually around 10 mg to 200 mg.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • the compounds can be administered in various modes, e.g. orally, topically, or by injection.
  • the precise amount of compound administered to a patient will be the responsibility of the attendant physician.
  • the specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diets, time of administration, route of administration, rate of excretion, drug combination, the precise disorder being treated, and the severity of the indication or condition being treated.
  • the route of administration may vary depending on the condition and its severity.
  • the compounds described herein may be administered in combination with another therapeutic agent.
  • another therapeutic agent such as a pharmaceutically acceptable salt, ester, or prodrug thereof.
  • the therapeutic effectiveness of one of the compounds described herein may be enhanced by administration of an adjuvant (i.e., by itself the adjuvant may only have minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced).
  • the benefit of experienced by a patient may be increased by administering one of the compounds described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit.
  • another therapeutic agent which also includes a therapeutic regimen
  • increased therapeutic benefit may result by also providing the patient with another therapeutic agent for diabetes.
  • the overall benefit experienced by the patient may simply be additive of the two therapeutic agents or the patient may experience a synergistic benefit.
  • the multiple therapeutic agents may be administered in any order or even simultaneously. If simultaneously, the multiple therapeutic agents may be provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills). One of the therapeutic agents may be given in multiple doses, or both may be given as multiple doses. If not simultaneous, the timing between the multiple doses may be any duration of time ranging from a few minutes to four weeks.
  • certain embodiments provide methods for treating NF- ⁇ B-mediated disorders in a human or animal subject in need of such treatment comprising administering to said subject an amount of a compound disclosed herein effective to reduce or prevent said disorder in the subject, in combination with at least one additional agent for the treatment of said disorder that is known in the art.
  • certain embodiments provide therapeutic compositions comprising at least one compound disclosed herein in combination with one or more additional agents for the treatment of NF- ⁇ B-mediated disorders.
  • Specific diseases to be treated by the compounds, compositions, and methods disclosed herein include ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, muscular dystrophy, muscle wasting, catabolic disorders, type 1 diabetes, type 2 diabetes, obesity, fetal growth retardation, hypercholesterolemia, atherosclerosis, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, chronic obstructive pulmonary disease, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, leptospiriosis renal disease, gut diseases, peritoneal endometriosis, skin diseases, nasal sinusitis, mesothelioma, anhidrotic ecodermal dysplasia-ID, behcet's disease, incontinentia pigmenti, tuberculosis, asthma, arthritis, crohn's disease, colitis, ocular allergy, gla
  • certain compounds and formulations disclosed herein may also be useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like. More preferred animals include horses, dogs, and cats.
  • Examples 6-7, 15, 20, and 33-34 can be synthesized according to scheme I.
  • Examples 8, 16-17, 26, and 35-36 can be synthesized according to scheme II.
  • Examples 11, 13, 18, 28, and 39-40 can be synthesized according to scheme IV.
  • Example 24 can be synthesized according to scheme V.
  • Examples 1-3, 25, and 29-32 can be synthesized according to scheme VI.
  • Preparation 1 can be synthesized according to scheme VII.
  • Example 10 can be synthesized according to scheme VIII.
  • the title compound can be synthesized according to the procedure of Example 8, Step 2, substituting 2-((10S,13S,17R)-17-hydroxy-10,13-dimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate for 2-oxo-2-((10S,13S,16R,17S)-10,13,16-trimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)ethyl acetate.
  • the reaction mixture is heated, maintained at 80 to 85° C. for 1 hour, and then cooled to room temperature.
  • Methanol (7 liters) is added.
  • the precipitated crystals are separated by filtration, washed with methanol and water, and dried under reduced pressure to give the title compound.
  • a pump was set up with methyl magnesium chloride (3M, 45.0 ml) and pumped into reactor 1 over 45 min (pump setting at 1.0 ml/min). Reactor 1 was stirred further at ⁇ 50° C. for 1 hour, then warmed to ⁇ 30° C. overnight.
  • Toluene (1 L) was added and the temperature brought to 0° C.
  • the mixture was extracted with 2 ⁇ 500 ml of 5% acetic acid (cold), then with 200 ml of 25% sodium chloride.
  • the aqueous phases were back extracted with 300 ml of toluene.
  • the combined toluene extracts were dried over magnesium sulfate, filtered, and concentrated to a viscous oil. Yield—57.8 grams.
  • 2-oxo-2-((10S,13S,16R,17S)-10,13,16,17-tetramethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)ethyl acetate A mixture of 2-((10S,13S)-10,13-dimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15-decahydro-1H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate (150 g) and copper propionate (1.9M in THF (90 ml) is cooled in an ice acetone bath.
  • Methyl magnesium chloride (1.96M in THF, 240 ml) is added dropwise for 30 min. After 1 hour, the reaction is quenched with methyl iodide (100 g) in 200 ml THF. The reaction mixture is then partitioned with water and toluene. The separated organic phase is washed with water, dried over sodium sulfate and concentrated. The residue is crystallized from ether and hexane to give the title compound.
  • C2C12 skeletal muscle cells stably transfected with a luciferase reporter construct regulated under multiple copies of the NF-kB response element (Panomics, Fremont, Calif.) were used to screen NF-kB inhibitors. These cells were maintained at 37° C. with 5% CO 2 in a tissue culture incubator with Dulbecco's modified Eagle medium (DMEM) medium containing 10% Fetal bovine serum (FBS) (ATCC, Manassas, Va.), Penicillin 100 U/ml, Streptomycin 100 ⁇ g/ml, and 100 ⁇ g/ml Hygromysin B (Roche, Indianapolis, Ind.).
  • DMEM Dulbecco's modified Eagle medium
  • FBS Fetal bovine serum
  • Penicillin 100 U/ml Streptomycin 100 ⁇ g/ml
  • Hygromysin B (Roche, Indianapolis, Ind.).
  • Screening assays were performed in myoblasts (grown in medium containing 10% FBS) in duplicate 96 well plates at a cell concentration of 5 ⁇ 10 4 cells per well in 100 ul volume. Cells were pretreated with various concentrations (0.01 ug/ml to 10 ug/ml) of compound for 24 hr duration before stimulating with tumor necrosis factor- ⁇ (TNF- ⁇ ) (10 ng/ml) for another 24 hrs. Prednisolone was included in every plate tested as a positive control.
  • TNF- ⁇ tumor necrosis factor- ⁇
  • Some of the compounds disclosed herein were tested in the C2C12 skeletal muscle cell luciferase assay and exhibited ⁇ 100% inhibition at concentrations of 0.01, 0.1, and 1 ug/mL; 80-100% inhibition at concentrations of 0.01, 0.1, 1, and 10 ug/mL; 60-80% inhibition at concentrations of 0.01, 0.1, and 1 ug/mL; 40-60% inhibition at concentrations of 1 and 10 ug/mL; and 20-40% inhibition at concentrations of 10 ug/mL.
  • TNF- ⁇ induced NF-kB activation was confirmed by nuclear translocation immunofluorescence assay.
  • C2C12 cells were grown on cover slips and treated with TNF- ⁇ and compound at optimal concentrations as described above. Cells were fixed with acetone and stained with a rabbit anti-NF-kB (p60) antibody/anti-rabbit Texas red (Santa Cruz Biotech, Inc, Santa Cruz, and CA) and counterstained with 4′,6-Diamidino-2-phenylindole HCl (DAPI) (Invitrogen, CA) to visualize the nuclei.
  • DAPI 4′,6-Diamidino-2-phenylindole HCl
  • Prednisolone treated mice showed significantly lower body weight (P ⁇ 0.05) than untreated mice at 33.8 weeks age.
  • Mice treated with some of the compounds disclosed herein at 20 and 40 mg/kg/day dosages gained significantly more body weight and gastrocnemius muscle mass than untreated mice.
  • Rota-rod Ugo Basile, VA, Italy
  • mice were trained on the rota-rod for two days before collecting data.
  • Each acclimatization session consisted of four training sessions, 2 per day and each session lasting 120 seconds at a speed of 5 rpm).
  • Each trial consisted of placing the mice on the rod at 10 rpm for 60 seconds (stabilizing period) followed by an acceleration from 10 rpm to 40 rpm within the first 25 seconds until the animal fell from the rod or until 180 seconds are reached. If the animals fell during the stabilizing period, they were placed back on the rod to complete the session.
  • the total testing time was 240 seconds (60 seconds stabilization time and 180 seconds test time).
  • mice treated with some of the compounds disclosed herein at 20 and 40 mg/kg/day dosages showed increased latency to fall at 12, 24, or 36 weeks. In some instances, latency to fall was decreased or unchanged, increased by 0-10%, increased by 10-20%, increased by 20-30%, or increased by 30-40%.
  • the distal tendon of the extensor digitorum longus (EDL) muscle was tied securely to the lever arm of a servomotor/force transducer (model 305B, Aurora Scientific, Richmond Hill, ON, Canada) and the proximal tendon to a tissue clamp. Muscles were stimulated between two platinum electrodes. With supramaximal stimulation of the muscle using single 0.2-ms square stimulation pulses for the EDL, muscle length was adjusted to the length (L o ) that resulted in maximal twitch force.
  • the maximum isometric tetanic force force (P o ) developed during a 300 ms train of stimulation pulses was recorded for the EDL muscle.
  • the muscle length was then measured with calipers and after removal of the muscle from the bath the mass of the muscle was determined.
  • the optimum fiber length (L f ) was calculated by multiplying L o by a previously determined L f /L o ratio of 0.45.
  • Total muscle fiber cross-sectional area was determined by dividing the wet mass by the product of L f and the density of mammalian skeletal muscle (1.06 mg/mm 3 ).
  • isometric specific force was determined by dividing P o by the total muscle fiber cross-sectional area. There was no statistically significant (P ⁇ 0.05) difference in specific force between untreated and prednisolone groups. Mice treated with some of the compounds disclosed herein at 20 and 40 mg/kg/day dosages showed statistically significant (P ⁇ 0.05) increased isometric specific force. In some instances, isometric specific force was decreased or unchanged, increased by 0-5%, increased by 5-10%, increased by 10-15%, or increased by 15-20%.
  • Hematoxylin and Eosin staining of gastrocnemius muscle of untreated mdx mice show significant degeneration and inflammation.
  • Skeletal muscle from Example 1- and Example 2-treated mice showed significant decrease in inflammation, degeneration, and increase in regenerating muscle fibers in comparison to untreated and prednisolone treated mdx mice.
  • Continuous administration of prednisolone appeared to increase degeneration and decrease in regeneration of dystrophic skeletal muscle.
  • a ligand binding assay was performed using cDNA expression clones (Baculovirus) for human and mouse glucocorticoid receptor-alpha. Liver extracts containing different GR constructs were incubated with radiolabeled 3H-Dexamethsone (Amersham Pharmacia Biotech) and test compound in assay buffer (10 mM Tris-HCl, 1.5 mM EDTA, 10% glycerol, 1 mM dithiothreitol, and 20 mM sodium molybdate, pH 7.6). The amount of radioactivity was measured using a scintillation plate reader.
  • Dexamethasone showed competitive binding with 3H-dexamethasone at micromolar concentrations. Some of the compounds disclosed herein were tested in the glucocorticoid receptor binding assay and found to have no significant (>75%) competitive binding to the glucocorticoid receptor at millimolar concentrations.

Abstract

The present invention relates to compounds and methods which may be useful as treatments of neuromuscular diseases such as muscular dystrophy, and as inhibitors of NF-κB for the treatment or prevention of muscular wasting disease, including muscular dystrophy.

Description

This application claims the benefit of priority of U.S. provisional application No. 61/320,905, filed Apr. 5, 2010, the disclosure of which is hereby incorporated by reference as if written herein in its entirety.
This invention was made with support from the U.S. Government under Contract No. DOD 05118004 and NIH No. 1U54HD053177-01A1. The U.S. Government has certain rights to this invention.
Disclosed herein are new non-hormonal steroid compounds and compositions and their application as pharmaceuticals for the treatment of disease. Methods of modulation of NF-κB activity in a human or animal subject are also provided for the treatment of diseases mediated by NF-κB.
Muscular wasting diseases, such as muscular dystrophies, are a group of degenerative diseases that culminate in progressive skeletal muscle wasting leading to muscle weakness, a high incidence of bone fracture, wheelchair dependence, and in some cases death. Of the muscular dystrophies, Duchenne muscular dystrophy is the most severe and most widely recognized. Another muscular wasting disease which shows similar symptoms, although less severe than Duchenne muscular dystrophy, is Becker muscular dystrophy. Even though the defective dystrophin gene causing both Duchenne muscular dystrophy and Becker muscular dystrophy has been known for over 20 years, a cure is still lacking.
Several catabolic factors that act to destroy host tissues during the cachectic process have been identified. It appears that the oversecretion of inflammatory cytokines, specifically tumor necrosis factor-alpha (TNF-α), is one of the most likely causes of cachexia. Specifically, TNF-α can mimic most of the abnormalities that occur in cachexia such as weight loss, anorexia, increased thermogenesis, changes in lipid metabolism, insulin resistance, and muscle wasting.
Muscle atrophy can also be induced by the loss of innervation or damage to innervation of the muscle tissue. Diseases such as chronic neuropathy and motor neuron disease can cause damage to innervation. Physical injury to the nerve can also lead to damage to the innervation of the muscle tissue. Alternatively, muscle atrophy can be the result of environmental conditions such as during spaceflight or as a result of aging or extended bed rest. Under these environmental conditions, the muscles do not bear the usual weight load, resulting in muscle atrophy from disuse. Specifically, during muscle disuse, intracellular processes are activated to induce proteolysis, mainly through the ATP dependent ubiquitin proteasome pathway, which regulates the NF-κB pathway.
NF-κB is known to mediate extracellular signals responsible for the induction of genes involved in pro-inflammatory responses. NF-κB is sequestered in the cytoplasm of most non-stimulated cells through a non-covalent interaction with one of several proteins known as inhibitors of kappa-beta (IκBs) (May & Ghosh, (1997) Semin Cancer. Biol. 8, 63-73; May & Ghosh, (1998) Immunol. Today 19, 80-88; Ghosh et al., (1998) Annu. Rev. Immunol. 16, 225-260). Cellular stimuli associated with pro-inflammatory responses such as TNF-α activate kinases, which in turn activate NF-κB by phosphorylating IκBs. The kinases that phosphorylate IκBs are called IκB kinases (IKKs).
Phosphorylation targets IκBs for subsequent ubiquitination and degradation. This degradation of IκBs reveals the nuclear localization signal on NF-κB, allowing nuclear accumulation of activation, which leads to binding of DNA and control of specific gene expression. Phosphorylation of IκB is therefore an important step in the regulation of NF-κB downstream of many stimuli, although other mechanisms can lead to the activation of functional NF-κB.
The identification and characterization of kinases that phosphorylate IκBs has led to a better understanding of signaling pathways involving NF-κB activation. Several different subtypes of IKK have been identified thus far. IKKα was initially identified as an IκB kinase induced by TNF-α stimulation in HeLa cells (DiDonato et al., (1997) Nature 388, 548-554). Another IκB kinase homologous to IKKα was identified, termed IKKβ, and determined to be the major IκB kinase induced following TNFα stimulation (Takeda et al., (1999) Science 284, 313-316; U.S. Pat. No. 6,030,834, issued to Pots et al. (2000); U.S. Pat. No. 5,939,302, issued to Woronicz et al. (1999)). IKKα and IKKβ have an overall homology of 52% and a 65% homology in the kinase domain (Zandi et al., (1997) Cell 91, 243-252).
IκB protein kinases (IKKs) phosphorylate IκBs at specific serine residues. Specifically, they phosphorylate serines 32 and 36 of IκBζ (Traenckner et al., (1995) EMBO J. 14, 2876-2883; DiDonato et al., (1996) Mol. Cell. Biol. 16, 1295-1304). Phosphorylation of both sites is required to efficiently target IκBα for degradation. Furthermore, activation of IKKα and IκKβ is usually in response to NF-κB activating agents including phorbol 12-myristate 13-acetate (PMA), lipopolysaccharide (LPS), interleukin-1 (IL-1), TNF-α, reactive oxygen species, and DNA damaging agents. Mutant IKKα and IKKβ, which are catalytically inactive, can be used to block NF-κB stimulation. IκB kinases are therefore essential in the regulation of NF-κB activation processes downstream of inflammatory stimuli. In other pathways, IκB kinases may not be important.
IKKα and IKKβ have distinct structural motifs including an amino terminal serine-threonine kinase domain separated from a carboxyl proximal helix-loop-helix domain by a leucine zipper domain. These structural characteristics are unlike other kinases, and the non-catalytic domains are thought to be involved in protein-protein interactions. As such, proteins which bind to IKKs should be capable of regulating the activity of NF-κB and potentially regulating downstream events such as induction of NF-κB. For instance, NEMO (NF-κB Essential Modulator) is a protein which has been identified to bind to IKKs and facilitate kinase activity (Yamaoke et al., (1998) Cell 93, 1231-1240; Rothwarf et al., (1998) Nature 395, 287-300).
In vivo studies have shown that chronic NF-κB activation is associated with muscular wasting diseases such as Duchenne muscular dystrophy, and is further illustrated in US 2007/0225315 (Mar. 15, 2007). Specifically, muscle wasting was largely prevented in subjects that were heterozygous for the p65/RelA NF-κB subunit. An injection of an NF-κB activation inhibitor peptide was found to inhibit the dystrophic phenotype in affected mice subjects. Without being bound by a particular theory, it appears that chronic activation of NF-κB is required for the muscle wasting symptoms of Duchenne muscular dystrophy. As such, a drug-based therapy targeting NF-κB can be an effective strategy to treat Duchenne muscular dystrophy, as well as other forms of muscular wasting diseases.
In general, muscular wasting diseases may be treated in accordance with the present disclosure with a direct or indirect modulator of NF-κB. Indirect modulators of NF-κB include, for example, inhibitors of IκB kinases (IKKs) such as IKKα inhibitors and IKKβ inhibitors, and inhibitors functioning directly upstream from IKKs in the signaling pathway such as inhibitors of phosphoinositide dependent kinase (PDK) and inhibitors of Akt (also referred to as PKB).
As noted above, one suitable approach for modulating the NF-κB pathway is by binding to one of the IκB protein kinases (IKKs). By binding the IKKs, phosphorylation of IκBs is blocked and NF-κB cannot be activated. In one embodiment, direct inhibiting compounds of IKK catalytic activity can be administered for the purpose of blocking the NF-κB pathway and inhibiting a muscular wasting disease. Specifically, inhibitors of IKKα or their enantiomers, analogs, prodrugs, active metabolites, salts, and/or hydrates thereof can be administered to the subject for the purpose of inhibiting a muscular wasting disease.
Novel compounds and pharmaceutical compositions, certain of which have been found to modulate NF-κB have been discovered, together with methods of synthesizing and using the compounds including methods for the treatment of NF-κB-mediated diseases in a patient by administering the compounds.
In certain embodiments, disclosed herein are compounds having structural Formula I:
Figure US09198921-20151201-C00001

or a salt thereof, wherein:
said dashed line indicates an optional double bond;
R1, R2, R3, and R4 are each independently selected from the group consisting of hydrogen, unsubstituted lower alkyl, lower haloalkyl, and halogen;
R5 is selected from the group consisting of hydrogen, lower alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl, said lower alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkynyl, amido, amino, aryl, aryloxy, cycloalkyl, haloalkoxy, haloalkyl, heteroalkyl, heteroaryl, hydroxy, perhaloalkoxy, and thiol;
R6 is selected from the group consisting of hydrogen, hydroxyl, and lower alkyl, said lower alkyl being optionally substituted with one or more substituents selected from the group consisting of alkenyl, alkoxy, alkyl, alkynyl, aryl, aryloxy, haloalkoxy, haloalkyl, heteroalkyl, hydroxy, and thiol;
R7 and R8 are independently selected from the groups consisting of hydrogen, unsubstituted C1-3 alkyl, or R7 and R8 can be taken together to form oxo or C3-6 saturated cycloalkyl; and
R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkylamino, alkylthio, alkynyl, amido, amino, aryl, aryloxy, aroyl, carbamate, carboxyl, cyano, cycloalkyl, halogen, haloalkoxy, haloalkyl, heteroalkyl, heterocycloalkyl, heteroaryl, hydrazinyl, hydroxy, mercaptyl, nitro, oxo, perhaloalkoxy, sulfonate, alkylsulfonyl, N-sulfonamido, S-sulfonamido, and thiol.
In further embodiments, R7 and R8 are independently selected from the groups consisting of hydrogen, unsubstituted C2-C3 alkyl, or R7 and R8 can be taken together to form C3-6 saturated cycloalkyl; R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkylamino, alkylthio, alkynyl, amino, aryl, aryloxy, aroyl, carbamate, cyano, cycloalkyl, halogen, haloalkoxy, haloalkyl, heteroalkyl, heterocycloalkyl, heteroaryl, hydrazinyl, hydroxy, mercaptyl, perhaloalkoxy, sulfonate, alkylsulfonyl, N-sulfonamido, S-sulfonamido, and thiol: if R1 is hydrogen, methyl, —CH2F, or fluoro, R2, R3, R4, R5, R7, and R8 are each hydrogen, and R6 is hydroxyl, then R9 is not hydrogen, formyl, unsubstituted C1-C5 alkylacyl, or benzoyl; if R1 is hydrogen, methyl, —CH2F, or fluoro, R2, R3, R4, R7, and R8 are each hydrogen, R5 is methyl, and R6 is hydroxyl, then R9 is not hydrogen, formyl, unsubstituted C1-C5 alkylacyl, trifluoroacetyl, —C(O)-adamantyl, or benzoyl; if R1 is hydrogen, methyl, fluoro, or chloro, R2, R3, R4, R6, R7, and R8 are each hydrogen, and R5 is methyl, then R9 is not hydrogen, unsubstituted C1-C5 alkylacyl, or benzoyl; if said dashed line indicates a double bond, R1, R2, R3, R4, R7, and R8 are each hydrogen, and R5 and R6 are each methyl, then R9 is not hydrogen, acetyl, or benzoyl; if said dashed line indicates a double bond, R1, R2, R3, R4, R5, R7, and R8 are each hydrogen, and R6 is ethyl, then R9 is not acetyl; if said dashed line does not indicate a double bond, R1 is hydrogen or fluoro, and R2, R3, R4, R5, R6, R7, and R8 are each hydrogen, then R9 is not hydrogen or acetyl; if R1, R2, R4, R5, R7, and R8 are each hydrogen, R3 is methyl, and R6 is hydroxyl, then R9 is not hydrogen, formyl, unsubstituted C1-C5 alkylacyl, or benzoyl; and if R1 and R2 are each fluoro, R3, R4, R7, and R8 are each hydrogen, R5 is methyl, and R6 is hydroxyl, then R9 is not acetyl.
In further embodiments, R5 is selected from the group consisting of C2-C8 alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl, said C2-C8 alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkynyl, amido, amino, aryl, aryloxy, cycloalkyl, haloalkoxy, haloalkyl, heteroalkyl, heteroaryl, hydroxy, perhaloalkoxy, and thiol.
In further embodiments, R1 and R3 are each hydrogen; R2 and R4 are each independently selected from the group consisting of hydrogen, methyl, and fluorine; R5 is selected from the group consisting of hydrogen, unsubstituted lower alkyl, and phenyl; R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; R7 and R8 are each hydrogen; and R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino, hydroxyl, and carboxyl.
In further embodiments, R2 and R4 are each hydrogen; R5 is selected from the group consisting of hydrogen, unsubstituted lower alkyl, and phenyl; R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; R7 and R8 are each hydrogen; and R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino, hydroxyl, and carboxyl.
In further embodiments, R5 is selected from the group consisting of hydrogen, methyl, and ethyl; R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; and R9 is selected from the group consisting of hydrogen, acetyl, and —C(O)CH2CH2CO2H.
In further embodiments, R5 is selected from the group consisting of unsubstituted C2-C6 alkyl and phenyl; R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; and R9 is selected from the group consisting of hydrogen, acetyl, and —C(O)CH2CH2CO2H.
In further embodiments, R5 is ethyl.
In further embodiments, said dashed line indicates an optional double bond; R1, R2, R3, and R4 are each independently selected from the group consisting of hydrogen, unsubstituted lower alkyl, lower haloalkyl, and halogen; R5 is selected from the group consisting of hydrogen, lower alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl, said lower alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkynyl, amido, amino, aryl, aryloxy, cycloalkyl, haloalkoxy, haloalkyl, heteroalkyl, heteroaryl, hydroxy, perhaloalkoxy, and thiol; R6 is selected from the group consisting of hydrogen, hydroxyl, and lower alkyl, said lower alkyl being optionally substituted with one or more substituents selected from the group consisting of alkenyl, alkoxy, alkyl, alkynyl, aryl, aryloxy, haloalkoxy, haloalkyl, heteroalkyl, hydroxy, and thiol; R7 and R8 are independently selected from the groups consisting of hydrogen, unsubstituted C1-3 alkyl, or R7 and R8 can be taken together to form oxo or C3-6 saturated cycloalkyl; and R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkylamino, alkylthio, alkynyl, amido, amino, aryl, aryloxy, aroyl, carbamate, carboxyl, cyano, cycloalkyl, halogen, haloalkoxy, haloalkyl, heteroalkyl, heterocycloalkyl, heteroaryl, hydrazinyl, hydroxy, mercaptyl, nitro, oxo, perhaloalkoxy, alkylsulfonyl, N-sulfonamido, S-sulfonamido, and thiol; if R1, R2, R3, R4, R7, and R8 are each hydrogen, R5 is methyl, and R6 is hydroxyl, then R9 is not —C(O)CH2CH2CO2H; if said dashed line does not indicate a double bond, R1, R2, R3, R4, R5, R7, and R8 are each hydrogen, and R6 is hydroxyl, then R9 is not —C(O)CH2CH2CO2H; if said dashed line indicates a double bond, R1 is methyl, R2, R3, R4, R5, R7, and R8 are each hydrogen, and R6 is hydroxyl, then R9 is not —C(O)CH2CH2CO2H; if R1, R2, R3, R4, R5, R7, and R8 are each hydrogen and R6 is hydroxyl, then R9 is not hydrogen or acetyl; if said dashed line does not indicate a double bond, R1 is fluorine, R2, R3, R4, R7, and R8 are each hydrogen, R5 is methyl, and R6 is hydroxyl, then R9 is not hydrogen or acetyl; and if said dashed line does not indicate a double bond, R1 is methyl, R2, R3, R4, R5, R7, and R8 are each hydrogen, and R6 is hydroxyl, then R9 is not acetyl.
In further embodiments, R7 and R8 are independently selected from the groups consisting of hydrogen, unsubstituted C2-C3 alkyl, or R7 and R8 can be taken together to form C3-6 saturated cycloalkyl; R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkylamino, alkylthio, alkynyl, amino, aryl, aryloxy, aroyl, carbamate, cyano, cycloalkyl, halogen, haloalkoxy, haloalkyl, heteroalkyl, heterocycloalkyl, heteroaryl, hydrazinyl, hydroxy, mercaptyl, perhaloalkoxy, alkylsulfonyl, N-sulfonamido, S-sulfonamido, and thiol; if R1 is hydrogen, methyl, —CH2F, or fluoro, R2, R3, R4, R5, R7, and R8 are each hydrogen, and R6 is hydroxyl, then R9 is not hydrogen, formyl, unsubstituted C1-C5 alkylacyl, or benzoyl; if R1 is hydrogen, methyl, —CH2F, or fluoro, R2, R3, R4, R7, and R8 are each hydrogen, R5 is methyl, and R6 is hydroxyl, then R9 is not hydrogen, formyl, unsubstituted C1-C5 alkylacyl, trifluoroacetyl, —C(O)-adamantyl, or benzoyl; if R1 is hydrogen, methyl, fluoro, or chloro, R2, R3, R4, R6, R7, and R8 are each hydrogen, and R5 is methyl, then R9 is not hydrogen, unsubstituted C1-C5 alkylacyl, or benzoyl; if said dashed line indicates a double bond, R1, R2, R3, R4, R7, and R8 are each hydrogen, and R5 and R6 are each methyl, then R9 is not hydrogen, acetyl, or benzoyl; if said dashed line indicates a double bond, R1, R2, R3, R4, R5, R7, and R8 are each hydrogen, and R6 is ethyl, then R9 is not acetyl; if said dashed line does not indicate a double bond, R1 is hydrogen or fluoro, and R2, R3, R4, R5, R6, R7, and R8 are each hydrogen, then R9 is not hydrogen or acetyl; if R1, R2, R4, R5, R7, and R8 are each hydrogen, R3 is methyl, and R6 is hydroxyl, then R9 is not hydrogen, formyl, unsubstituted C1-C5 alkylacyl, or benzoyl; and if R1 and R2 are each fluoro, R3, R4, R7, and R8 are each hydrogen, R5 is methyl, and R6 is hydroxyl, then R9 is not acetyl.
In further embodiments, R5 is selected from the group consisting of C2-C8 alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl, said C2-C8 alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkynyl, amido, amino, aryl, aryloxy, cycloalkyl, haloalkoxy, haloalkyl, heteroalkyl, heteroaryl, hydroxy, perhaloalkoxy, and thiol.
In further embodiments, R1 and R3 are each hydrogen; R2 and R4 are each independently selected from the group consisting of hydrogen, methyl, and fluorine; R5 is selected from the group consisting of hydrogen, unsubstituted lower alkyl, and phenyl; R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; R7 and R8 are each hydrogen; and R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino, hydroxyl, and carboxyl.
In further embodiments, R2 and R4 are each hydrogen; R5 is selected from the group consisting of hydrogen, unsubstituted lower alkyl, and phenyl; R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; R7 and R8 are each hydrogen; and R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino, hydroxyl, and carboxyl.
In further embodiments, R5 is selected from the group consisting of hydrogen, methyl, and ethyl; R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; and R9 is selected from the group consisting of hydrogen, acetyl, and —C(O)CH2CH2CO2H.
In further embodiments, R5 is selected from the group consisting of unsubstituted C2-C6 alkyl and phenyl; R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; and R9 is selected from the group consisting of hydrogen, acetyl, and —C(O)CH2CH2CO2H.
In further embodiments, R5 is ethyl.
In further embodiments, said dashed line indicates an optional double bond; R1, R2, R3, and R4 are each independently selected from the group consisting of hydrogen, unsubstituted lower alkyl, lower haloalkyl, and halogen; R5 is selected from the group consisting of hydrogen, lower alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl, said lower alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkynyl, amido, amino, aryl, aryloxy, cycloalkyl, haloalkoxy, haloalkyl, heteroalkyl, heteroaryl, hydroxy, perhaloalkoxy, and thiol; R6 is selected from the group consisting of hydrogen, hydroxyl, and lower alkyl, said lower alkyl being optionally substituted with one or more substituents selected from the group consisting of alkenyl, alkoxy, alkyl, alkynyl, aryl, aryloxy, haloalkoxy, haloalkyl, heteroalkyl, hydroxy, and thiol; R7 and R8 are independently selected from the groups consisting of hydrogen, unsubstituted C2-C3 alkyl, or R7 and R8 can be taken together to form C3-6 saturated cycloalkyl; R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkylamino, alkylthio, alkynyl, amino, aryl, aryloxy, aroyl, carbamate, cyano, cycloalkyl, halogen, haloalkoxy, haloalkyl, heteroalkyl, heterocycloalkyl, heteroaryl, hydrazinyl, hydroxy, mercaptyl, perhaloalkoxy, sulfonate, alkylsulfonyl, N-sulfonamido, S-sulfonamido, and thiol; if R1 is hydrogen, methyl, —CH2F, or fluoro, R2, R3, R4, R5, R7, and R8 are each hydrogen, and R6 is hydroxyl, then R9 is not hydrogen, formyl, unsubstituted C1-C5 alkylacyl, or benzoyl; if R1 is hydrogen, methyl, —CH2F, or fluoro, R2, R3, R4, R7, and R8 are each hydrogen, R5 is methyl, and R6 is hydroxyl, then R9 is not hydrogen, formyl, unsubstituted C1-C5 alkylacyl, trifluoroacetyl, —C(O)-adamantyl, or benzoyl; if R1 is hydrogen, methyl, fluoro, or chloro, R2, R3, R4, R6, R7, and R8 are each hydrogen, and R5 is methyl, then R9 is not hydrogen, unsubstituted C1-C5 alkylacyl, or benzoyl; if said dashed line indicates a double bond, R1, R2, R3, R4, R7, and R8 are each hydrogen, and R5 and R6 are each methyl, then R9 is not hydrogen, acetyl, or benzoyl; if said dashed line indicates a double bond, R1, R2, R3, R4, R5, R7, and R8 are each hydrogen, and R6 is ethyl, then R9 is not acetyl; if said dashed line does not indicate a double bond, R1 is hydrogen or fluoro, and R2, R3, R4, R5, R6, R7, and R8 are each hydrogen, then R9 is not hydrogen or acetyl; if R1, R2, R4, R5, R7, and R8 are each hydrogen, R3 is methyl, and R6 is hydroxyl, then R9 is not hydrogen, formyl, unsubstituted C1-C5 alkylacyl, or benzoyl; and if R1 and R2 are each fluoro, R3, R4, R7, and R8 are each hydrogen, R5 is methyl, and R6 is hydroxyl, then R9 is not acetyl.
In further embodiments, R1 and R3 are each hydrogen; R2 and R4 are each independently selected from the group consisting of hydrogen, methyl, and fluorine; R5 is selected from the group consisting of hydrogen, unsubstituted lower alkyl, and phenyl; R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; R7 and R8 are each hydrogen; and R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino and hydroxyl.
In further embodiments, R2 and R4 are each hydrogen; R5 is selected from the group consisting of hydrogen, unsubstituted lower alkyl, and phenyl; R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; R7 and R8 are each hydrogen; and R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino and hydroxyl.
In further embodiments, R5 is selected from the group consisting of hydrogen, methyl, and ethyl; R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; and R9 is selected from the group consisting of hydrogen and acetyl.
In further embodiments, said dashed line indicates an optional double bond; R1, R2, R3, and R4 are each independently selected from the group consisting of hydrogen, unsubstituted lower alkyl, lower haloalkyl, and halogen; R5 is selected from the group consisting of C2-C8 alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl, said C2-C8 alkyl, aryl, cycloalkyl, heterocycloalkyl, and heteroaryl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkynyl, amido, amino, aryl, aryloxy, cycloalkyl, haloalkoxy, haloalkyl, heteroalkyl, heteroaryl, hydroxy, perhaloalkoxy, and thiol; R6 is selected from the group consisting of hydrogen, hydroxyl, and lower alkyl, said lower alkyl being optionally substituted with one or more substituents selected from the group consisting of alkenyl, alkoxy, alkyl, alkynyl, aryl, aryloxy, haloalkoxy, haloalkyl, heteroalkyl, hydroxy, and thiol; R7 and R8 are independently selected from the groups consisting of hydrogen, unsubstituted C1-3 alkyl, or R7 and R8 can be taken together to form oxo or C3-6 saturated cycloalkyl; and R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with one or more substituents selected from the group consisting of acyl, alkenyl, alkoxy, alkyl, alkylamino, alkylthio, alkynyl, amido, amino, aryl, aryloxy, aroyl, carbamate, carboxyl, cyano, cycloalkyl, halogen, haloalkoxy, haloalkyl, heteroalkyl, heterocycloalkyl, heteroaryl, hydrazinyl, hydroxy, mercaptyl, nitro, oxo, perhaloalkoxy, sulfonate, alkylsulfonyl, N-sulfonamido, S-sulfonamido, and thiol.
In further embodiments, R1 and R3 are each hydrogen; R2 and R4 are each independently selected from the group consisting of hydrogen, methyl, and fluorine; R5 is selected from the group consisting of unsubstituted C2-C6 alkyl and phenyl; R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; R7 and R8 are each hydrogen; and R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino, hydroxyl, and carboxyl.
In further embodiments, R2 and R4 are each hydrogen; R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; R7 and R8 are each hydrogen; and R9 is selected from the group consisting of hydrogen, acyl, and alkyl, said acyl and alkyl being optionally substituted with amino, hydroxyl, and carboxyl.
In further embodiments, R6 is selected from the group consisting of hydrogen, hydroxyl, and methyl; and R9 is selected from the group consisting of hydrogen, acetyl, and —C(O)CH2CH2CO2H.
In further embodiments, R5 is ethyl.
Certain compounds disclosed herein may possess useful NF-κB modulating activity, and may be used in the treatment or prophylaxis of a disease or condition in which NF-κB plays an active role. Thus, in broad aspect, certain embodiments also provide pharmaceutical compositions comprising one or more compounds disclosed herein together with a pharmaceutically acceptable carrier, as well as methods of making and using the compounds and compositions. Certain embodiments provide methods for modulating NF-κB. Other embodiments provide methods for treating a NF-κB-mediated disorder in a patient in need of such treatment, comprising administering to said patient a therapeutically effective amount of a compound or composition according to the present invention. Also provided is the use of certain compounds disclosed herein for use in the manufacture of a medicament for the treatment of a disease or condition ameliorated by the modulation of NF-κB.
In certain embodiments, said NF-κB-mediated disease is selected from the group consisting of muscular dystrophy, arthritis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type 2 diabetes, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, muscle wasting, catabolic disorders, obesity, fetal growth retardation, hypercholesterolemia, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, asthma, chronic obstructive pulmonary disease, Sjogren's syndrome, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, gut diseases, peritoneal endometriosis, skin diseases, nasal sinusitis, mesothelioma, anhidrotic ecodermal dysplasia-ID, behcet's disease, incontinentia pigmenti, tuberculosis, asthma, crohn's disease, colitis, ocular allergy, appendicitis, paget's disease, pancreatitis, periodonitis, endometriosis, inflammatory bowel disease, inflammatory lung disease, silica-induced diseases, sleep apnea, AIDS, HIV-1, autoimmune diseases, antiphospholipid syndrome, lupus, lupus nephritis, familial mediterranean fever, hereditary periodic fever syndrome, psychosocial stress diseases, neuropathological diseases, familial amyloidotic polyneuropathy, inflammatory neuropathy, parkinson's disease, multiple sclerosis, alzheimer's disease, amyotropic lateral sclerosis, huntington's disease, cataracts, and hearing loss.
In further embodiments, said disease is said NF-κB-mediated disease is asthma or chronic obstructive pulmonary disease.
In further embodiments, said NF-κB-mediated is Sjogren's syndrome.
In further embodiments, said NF-κB-mediated is arthritis.
In further embodiments, said NF-κB-mediated is muscular wasting.
In further embodiments, said muscular wasting disease is muscular dystrophy.
In further embodiments, said muscular dystrophy is selected from the group consisting of Duchenne muscular dystrophy, Becker muscular dystrophy, limb girdle muscular dystrophy, congenital muscular dystrophy, facioscapulohumeral muscular dystrophy, myotonic muscular dystrophy, oculopharyngeal muscular dystrophy, distal muscular dystrophy, and Emery-Dreifuss muscular dystrophy.
In further embodiments, said muscular dystrophy is Duchenne muscular dystrophy.
In certain embodiments, disclosed herein is a compound selected from the group consisting of examples 25-40.
In certain embodiments, disclosed herein is a pharmaceutical composition comprising a compound selected from the group consisting of examples 25-40 together with a pharmaceutically acceptable carrier.
In certain embodiments, said pharmaceutical composition comprises a tablet or a capsule.
In certain embodiments, said pharmaceutical composition comprises another therapeutic agent.
In certain embodiments, disclosed herein is a method of reducing the symptoms of a disease selected from the group consisting of muscular dystrophy, arthritis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type 2 diabetes, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, muscle wasting, catabolic disorders, obesity, fetal growth retardation, hypercholesterolemia, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, asthma, chronic obstructive pulmonary disease, Sjogren's syndrome, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, gut diseases, peritoneal endometriosis, skin diseases, nasal sinusitis, mesothelioma, anhidrotic ecodermal dysplasia-ID, behcet's disease, incontinentia pigmenti, tuberculosis, asthma, crohn's disease, colitis, ocular allergy, appendicitis, paget's disease, pancreatitis, periodonitis, endometriosis, inflammatory bowel disease, inflammatory lung disease, silica-induced diseases, sleep apnea, AIDS, HIV-1, autoimmune diseases, antiphospholipid syndrome, lupus, lupus nephritis, familial mediterranean fever, hereditary periodic fever syndrome, psychosocial stress diseases, neuropathological diseases, familial amyloidotic polyneuropathy, inflammatory neuropathy, parkinson's disease, multiple sclerosis, alzheimer's disease, amyotropic lateral sclerosis, huntington's disease, cataracts, and hearing loss, comprising the administration, to a patient in need thereof, of a therapeutically effective amount of a compound selected from the group consisting of examples 25-40.
In certain embodiments, disclosed herein is a method of reducing the symptoms of a disease selected from the group consisting of muscular dystrophy, arthritis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type 2 diabetes, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, muscle wasting, catabolic disorders, obesity, fetal growth retardation, hypercholesterolemia, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, asthma, chronic obstructive pulmonary disease, Sjogren's syndrome, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, gut diseases, peritoneal endometriosis, skin diseases, nasal sinusitis, mesothelioma, anhidrotic ecodermal dysplasia-ID, behcet's disease, incontinentia pigmenti, tuberculosis, asthma, crohn's disease, colitis, ocular allergy, appendicitis, paget's disease, pancreatitis, periodonitis, endometriosis, inflammatory bowel disease, inflammatory lung disease, silica-induced diseases, sleep apnea, AIDS, HIV-1, autoimmune diseases, antiphospholipid syndrome, lupus, lupus nephritis, familial mediterranean fever, hereditary periodic fever syndrome, psychosocial stress diseases, neuropathological diseases, familial amyloidotic polyneuropathy, inflammatory neuropathy, parkinson's disease, multiple sclerosis, alzheimer's disease, amyotropic lateral sclerosis, huntington's disease, cataracts, and hearing loss, comprising the administration, to a patient in need thereof, of a therapeutically effective amount of a compound selected from the group consisting of examples 25-40.
In certain embodiments, said disease is muscular dystrophy.
In certain embodiments, said muscular dystrophy is selected from the group consisting of Duchenne muscular dystrophy, Becker muscular dystrophy, limb girdle muscular dystrophy, congenital muscular dystrophy, facioscapulohumeral muscular dystrophy, myotonic muscular dystrophy, oculopharyngeal muscular dystrophy, distal muscular dystrophy, and Emery-Dreifuss muscular dystrophy.
In certain embodiments, said muscular dystrophy is Duchenne muscular dystrophy.
In certain embodiments, said disease is selected from the group consisting of asthma and chronic obstructive pulmonary disease.
In certain embodiments, said disease is asthma.
In certain embodiments, said disease is chronic obstructive pulmonary disease.
In certain embodiments, said disease is arthritis.
In certain embodiments, said disease is selected from the group consisting of traumatic brain injury and spinal cord injury.
In certain embodiments, said disease is traumatic brain injury.
In certain embodiments, said disease is spinal cord injury.
In certain embodiments, said disease is amyotropic lateral sclerosis.
In certain embodiments, said disease is Sjogren's syndrome.
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00002
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00003
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00004
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00005
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00006
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00007
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00008
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00009
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00010
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00011
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00012
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00013
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00014
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00015
In certain embodiments, said compound has the structural formula:
Figure US09198921-20151201-C00016
In certain embodiments, said method comprises the administration of another therapeutic agent.
In certain embodiments, disclosed herein is a compound selected from the group consisting of examples 25-40 for use as a medicament.
In certain embodiments, disclosed herein is a compound selected from the group consisting of examples 25-40 for use in the manufacture of a medicament for the treatment of a disease selected from the group consisting of muscular dystrophy, arthritis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type 2 diabetes, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, muscle wasting, catabolic disorders, obesity, fetal growth retardation, hypercholesterolemia, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, asthma, chronic obstructive pulmonary disease, Sjogren's syndrome, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, gut diseases, peritoneal endometriosis, skin diseases, nasal sinusitis, mesothelioma, anhidrotic ecodermal dysplasia-ID, behcet's disease, incontinentia pigmenti, tuberculosis, asthma, crohn's disease, colitis, ocular allergy, appendicitis, paget's disease, pancreatitis, periodonitis, endometriosis, inflammatory bowel disease, inflammatory lung disease, silica-induced diseases, sleep apnea, AIDS, HIV-1, autoimmune diseases, antiphospholipid syndrome, lupus, lupus nephritis, familial mediterranean fever, hereditary periodic fever syndrome, psychosocial stress diseases, neuropathological diseases, familial amyloidotic polyneuropathy, inflammatory neuropathy, parkinson's disease, multiple sclerosis, alzheimer's disease, amyotropic lateral sclerosis, huntington's disease, cataracts, and hearing loss, comprising the administration, to a patient in need thereof, of a therapeutically effective amount of a compound selected from the group consisting of examples 25-40.
When ranges of values are disclosed, and the notation “from n1 . . . to n2” is used, where n1 and n2 are the numbers, then unless otherwise specified, this notation is intended to include the numbers themselves and the range between them. This range may be integral or continuous between and including the end values. By way of example, the range “from 2 to 6 carbons” is intended to include two, three, four, five, and six carbons, since carbons come in integer units. Compare, by way of example, the range “from 1 to 3 μM (micromolar),” which is intended to include 1 μM, 3 μM, and everything in between to any number of significant figures (e.g., 1.255 μM, 2.1 μM, 2.9999 μM, etc.).
The term “about,” as used herein, is intended to qualify the numerical values which it modifies, denoting such a value as variable within a margin of error. When no particular margin of error, such as a standard deviation to a mean value given in a chart or table of data, is recited, the term “about” should be understood to mean that range which would encompass the recited value and the range which would be included by rounding up or down to that figure as well, taking into account significant figures.
The term “acyl,” as used herein, alone or in combination, refers to a carbonyl attached to an alkenyl, alkyl, aryl, cycloalkyl, heteroaryl, heterocycle, or any other moiety were the atom attached to the carbonyl is carbon. An “acetyl” group refers to a —C(O)CH3 group. An “alkylcarbonyl” or “alkanoyl” group refers to an alkyl group attached to the parent molecular moiety through a carbonyl group. Examples of such groups include methylcarbonyl and ethylcarbonyl. Examples of acyl groups include formyl, alkanoyl and aroyl.
The term “alkenyl,” as used herein, alone or in combination, refers to a straight-chain or branched-chain hydrocarbon group having one or more double bonds and containing from 2 to 20 carbon atoms. In certain embodiments, said alkenyl will comprise from 2 to 6 carbon atoms. The term “alkenylene” refers to a carbon-carbon double bond system attached at two or more positions such as ethenylene [(—CH═CH—),(—C::C—)]. Examples of suitable alkenyl groups include ethenyl, propenyl, 2-methylpropenyl, 1,4-butadienyl and the like. Unless otherwise specified, the term “alkenyl” may include “alkenylene” groups.
The term “alkoxy,” as used herein, alone or in combination, refers to an alkyl ether group, wherein the term alkyl is as defined below. Examples of suitable alkyl ether groups include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, iso-butoxy, sec-butoxy, tert-butoxy, and the like.
The term “alkyl,” as used herein, alone or in combination, refers to a straight-chain or branched-chain alkyl group containing from 1 to 20 carbon atoms. In certain embodiments, said alkyl will comprise from 1 to 10 carbon atoms. In further embodiments, said alkyl will comprise from 1 to 6 carbon atoms. Alkyl groups may be optionally substituted as defined herein. Examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, iso-amyl, hexyl, octyl, noyl and the like. The term “alkylene,” as used herein, alone or in combination, refers to a saturated aliphatic group derived from a straight or branched chain saturated hydrocarbon attached at two or more positions, such as methylene (—CH2—). Unless otherwise specified, the term “alkyl” may include “alkylene” groups.
The term “alkylamino,” as used herein, alone or in combination, refers to an alkyl group attached to the parent molecular moiety through an amino group. Suitable alkylamino groups may be mono- or dialkylated, forming groups such as, for example, N-methylamino, N-ethylamino, N,N-dimethylamino, N,N-ethylmethylamino and the like.
The term “alkylidene,” as used herein, alone or in combination, refers to an alkenyl group in which one carbon atom of the carbon-carbon double bond belongs to the moiety to which the alkenyl group is attached.
The term “alkylthio,” as used herein, alone or in combination, refers to an alkyl thioether (R—S—) group wherein the term alkyl is as defined above and wherein the sulfur may be singly or doubly oxidized. Examples of suitable alkyl thioether groups include methylthio, ethylthio, n-propylthio, isopropylthio, n-butylthio, iso-butylthio, sec-butylthio, tert-butylthio, methanesulfonyl, ethanesulfinyl, and the like.
The term “alkynyl,” as used herein, alone or in combination, refers to a straight-chain or branched chain hydrocarbon group having one or more triple bonds and containing from 2 to 20 carbon atoms. In certain embodiments, said alkynyl comprises from 2 to 6 carbon atoms. In further embodiments, said alkynyl comprises from 2 to 4 carbon atoms. The term “alkynylene” refers to a carbon-carbon triple bond attached at two positions such as ethynylene (—C:::C—, —C≡C—). Examples of alkynyl groups include ethynyl, propynyl, hydroxypropynyl, butyn-1-yl, butyn-2-yl, pentyn-1-yl, 3-methylbutyn-1-yl, hexyn-2-yl, and the like. Unless otherwise specified, the term “alkynyl” may include “alkynylene” groups.
The terms “amido” and “carbamoyl,” as used herein, alone or in combination, refer to an amino group as described below attached to the parent molecular moiety through a carbonyl group, or vice versa. The term “C-amido” as used herein, alone or in combination, refers to a —C(═O)—NR2 group with R as defined herein. The term “N-amido” as used herein, alone or in combination, refers to a RC(═O)NH— group, with R as defined herein. The term “acylamino” as used herein, alone or in combination, embraces an acyl group attached to the parent moiety through an amino group. An example of an “acylamino” group is acetylamino (CH3C(O)NH—).
The term “amino,” as used herein, alone or in combination, refers to —NRR′, wherein R and R′ are independently selected from the group consisting of hydrogen, alkyl, acyl, heteroalkyl, aryl, cycloalkyl, heteroaryl, and heterocycloalkyl, any of which may themselves be optionally substituted. Additionally, R and R′ may combine to form heterocycloalkyl, either of which may be optionally substituted.
The term “aryl,” as used herein, alone or in combination, means a carbocyclic aromatic system containing one, two or three rings wherein such polycyclic ring systems are fused together. The term “aryl” embraces aromatic groups such as phenyl, naphthyl, anthracenyl, and phenanthryl.
The term “arylalkenyl” or “aralkenyl,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkenyl group.
The term “arylalkoxy” or “aralkoxy,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkoxy group.
The term “arylalkyl” or “aralkyl,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkyl group.
The term “arylalkynyl” or “aralkynyl,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkynyl group.
The term “arylalkanoyl” or “aralkanoyl” or “aroyl,” as used herein, alone or in combination, refers to an acyl group derived from an aryl-substituted alkanecarboxylic acid such as benzoyl, napthoyl, phenylacetyl, 3-phenylpropionyl (hydrocinnamoyl), 4-phenylbutyryl, (2-naphthyl)acetyl, 4-chlorohydrocinnamoyl, and the like.
The term aryloxy as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an oxy.
The terms “benzo” and “benz,” as used herein, alone or in combination, refer to the divalent group C6H4=derived from benzene. Examples include benzothiophene and benzimidazole.
The term “carbamate,” as used herein, alone or in combination, refers to an ester of carbamic acid (—NHCOO—) which may be attached to the parent molecular moiety from either the nitrogen or acid end, and which may be optionally substituted as defined herein.
The term “O-carbamyl” as used herein, alone or in combination, refers to a —OC(O)NRR′, group-with R and R′ as defined herein.
The term “N-carbamyl” as used herein, alone or in combination, refers to a ROC(O)NR′— group, with R and R′ as defined herein.
The term “carbonyl,” as used herein, when alone includes formyl [—C(O)H] and in combination is a —C(O)— group.
The term “carboxyl” or “carboxy,” as used herein, refers to —C(O)OH or the corresponding “carboxylate” anion, such as is in a carboxylic acid salt. An “O-carboxy” group refers to a RC(O)O— group, where R is as defined herein. A “C-carboxy” group refers to a —C(O)OR groups where R is as defined herein.
The term “cyano,” as used herein, alone or in combination, refers to —CN.
The term “cycloalkyl,” or, alternatively, “carbocycle,” as used herein, alone or in combination, refers to a saturated or partially saturated monocyclic, bicyclic or tricyclic alkyl group wherein each cyclic moiety contains from 3 to 12 carbon atom ring members and which may optionally be a benzo fused ring system which is optionally substituted as defined herein. In certain embodiments, said cycloalkyl will comprise from 3 to 7 carbon atoms. In certain embodiments, said cycloalkyl will comprise from 5 to 7 carbon atoms. Examples of such cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, tetrahydronapthyl, indanyl, octahydronaphthyl, 2,3-dihydro-1H-indenyl, adamantyl and the like. “Bicyclic” and “tricyclic” as used herein are intended to include both fused ring systems, such as decahydronaphthalene, octahydronaphthalene as well as the multicyclic (multicentered) saturated or partially unsaturated type. The latter type of isomer is exemplified in general by, bicyclo[1,1,1]pentane, camphor, adamantane, and bicyclo[3,2,1]octane.
The term “ester,” as used herein, alone or in combination, refers to a carboxy group bridging two moieties linked at carbon atoms.
The term “ether,” as used herein, alone or in combination, refers to an oxy group bridging two moieties linked at carbon atoms.
The term “halo,” or “halogen,” as used herein, alone or in combination, refers to fluorine, chlorine, bromine, or iodine.
The term “haloalkoxy,” as used herein, alone or in combination, refers to a haloalkyl group attached to the parent molecular moiety through an oxygen atom.
The term “haloalkyl,” as used herein, alone or in combination, refers to an alkyl group having the meaning as defined above wherein one or more hydrogens are replaced with a halogen. Specifically embraced are monohaloalkyl, dihaloalkyl and polyhaloalkyl groups. A monohaloalkyl group, for one example, may have an iodo, bromo, chloro or fluoro atom within the group. Dihalo and polyhaloalkyl groups may have two or more of the same halo atoms or a combination of different halo groups. Examples of haloalkyl groups include fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl, difluoropropyl, dichloroethyl and dichloropropyl. “Haloalkylene” refers to a haloalkyl group attached at two or more positions. Examples include fluoromethylene (—CFH—), difluoromethylene (—CF2—), chloromethylene (—CHCl—) and the like.
The term “heteroalkyl,” as used herein, alone or in combination, refers to a stable straight or branched chain, or cyclic hydrocarbon group, or combinations thereof, fully saturated or containing from 1 to 3 degrees of unsaturation, consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of O, N, and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized. The heteroatom(s) O, N and S may be placed at any interior position of the heteroalkyl group. Up to two heteroatoms may be consecutive, such as, for example, —CH2—NH—OCH3.
The term “heteroaryl,” as used herein, alone or in combination, refers to a 3 to 7 membered unsaturated heteromonocyclic ring, or a fused monocyclic, bicyclic, or tricyclic ring system in which at least one of the fused rings is aromatic, which contains at least one atom selected from the group consisting of O, S, and N. In certain embodiments, said heteroaryl will comprise from 5 to 7 carbon atoms. The term also embraces fused polycyclic groups wherein heterocyclic rings are fused with aryl rings, wherein heteroaryl rings are fused with other heteroaryl rings, wherein heteroaryl rings are fused with heterocycloalkyl rings, or wherein heteroaryl rings are fused with cycloalkyl rings. Examples of heteroaryl groups include pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, pyranyl, furyl, thienyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, thiadiazolyl, isothiazolyl, indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, quinoxalinyl, quinazolinyl, indazolyl, benzotriazolyl, benzodioxolyl, benzopyranyl, benzoxazolyl, benzoxadiazolyl, benzothiazolyl, benzothiadiazolyl, benzofuryl, benzothienyl, chromonyl, coumarinyl, benzopyranyl, tetrahydroquinolinyl, tetrazolopyridazinyl, tetrahydroisoquinolinyl, thienopyridinyl, furopyridinyl, pyrrolopyridinyl and the like. Exemplary tricyclic heterocyclic groups include carbazolyl, benzindolyl, phenanthrolinyl, dibenzofuranyl, acridinyl, phenanthridinyl, xanthenyl and the like.
The terms “heterocycloalkyl” and, interchangeably, “heterocycle,” as used herein, alone or in combination, each refer to a saturated, partially unsaturated, or fully unsaturated monocyclic, bicyclic, or tricyclic heterocyclic group containing at least one heteroatom as a ring member, wherein each said heteroatom may be independently selected from the group consisting of nitrogen, oxygen, and sulfur In certain embodiments, said heterocycloalkyl will comprise from 1 to 4 heteroatoms as ring members. In further embodiments, said heterocycloalkyl will comprise from 1 to 2 heteroatoms as ring members. In certain embodiments, said heterocycloalkyl will comprise from 3 to 8 ring members in each ring. In further embodiments, said heterocycloalkyl will comprise from 3 to 7 ring members in each ring. In yet further embodiments, said heterocycloalkyl will comprise from 5 to 6 ring members in each ring. “Heterocycloalkyl” and “heterocycle” are intended to include sulfones, sulfoxides, N-oxides of tertiary nitrogen ring members, and carbocyclic fused and benzo fused ring systems; additionally, both terms also include systems where a heterocycle ring is fused to an aryl group, as defined herein, or an additional heterocycle group. Examples of heterocycle groups include aziridinyl, azetidinyl, 1,3-benzodioxolyl, dihydroisoindolyl, dihydroisoquinolinyl, dihydrocinnolinyl, dihydrobenzodioxinyl, dihydro[1,3]oxazolo[4,5-b]pyridinyl, benzothiazolyl, dihydroindolyl, dihy-dropyridinyl, 1,3-dioxanyl, 1,4-dioxanyl, 1,3-dioxolanyl, isoindolinyl, morpholinyl, piperazinyl, pyrrolidinyl, tetrahydropyridinyl, piperidinyl, thiomorpholinyl, and the like. The heterocycle groups may be optionally substituted unless specifically prohibited.
The term “hydrazinyl” as used herein, alone or in combination, refers to two amino groups joined by a single bond, i.e., —N—N—.
The term “hydroxy,” as used herein, alone or in combination, refers to —OH.
The term “hydroxyalkyl,” as used herein, alone or in combination, refers to a hydroxy group attached to the parent molecular moiety through an alkyl group.
The term “imino,” as used herein, alone or in combination, refers to ═N—.
The term “iminohydroxy,” as used herein, alone or in combination, refers to ═N(OH) and ═N—O—.
The phrase “in the main chain” refers to the longest contiguous or adjacent chain of carbon atoms starting at the point of attachment of a group to the compounds of any one of the formulas disclosed herein.
The term “isocyanato” refers to a —NCO group.
The term “isothiocyanato” refers to a —NCS group.
The phrase “linear chain of atoms” refers to the longest straight chain of atoms independently selected from carbon, nitrogen, oxygen and sulfur.
The term “lower,” as used herein, alone or in a combination, where not otherwise specifically defined, means containing from 1 to and including 6 carbon atoms.
The term “lower aryl,” as used herein, alone or in combination, means phenyl or naphthyl, which may be optionally substituted as provided.
The term “lower heteroaryl,” as used herein, alone or in combination, means either 1) monocyclic heteroaryl comprising five or six ring members, of which between one and four said members may be heteroatoms selected from the group consisting of O, S, and N, or 2) bicyclic heteroaryl, wherein each of the fused rings comprises five or six ring members, comprising between them one to four heteroatoms selected from the group consisting of O, S, and N.
The term “lower cycloalkyl,” as used herein, alone or in combination, means a monocyclic cycloalkyl having between three and six ring members. Lower cycloalkyls may be unsaturated. Examples of lower cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
The term “lower heterocycloalkyl,” as used herein, alone or in combination, means a monocyclic heterocycloalkyl having between three and six ring members, of which between one and four may be heteroatoms selected from the group consisting of O, S, and N. Examples of lower heterocycloalkyls include pyrrolidinyl, imidazolidinyl, pyrazolidinyl, piperidinyl, piperazinyl, and morpholinyl. Lower heterocycloalkyls may be unsaturated.
The term “lower amino,” as used herein, alone or in combination, refers to —NRR′, wherein R and R′ are independently selected from the group consisting of hydrogen, lower alkyl, and lower heteroalkyl, any of which may be optionally substituted. Additionally, the R and R′ of a lower amino group may combine to form a five- or six-membered heterocycloalkyl, either of which may be optionally substituted.
The term “mercaptyl” as used herein, alone or in combination, refers to an RS— group, where R is as defined herein.
The term “nitro,” as used herein, alone or in combination, refers to —NO2.
The terms “oxy” or “oxa,” as used herein, alone or in combination, refer to —O—.
The term “oxo,” as used herein, alone or in combination, refers to ═O.
The term “perhaloalkoxy” refers to an alkoxy group where all of the hydrogen atoms are replaced by halogen atoms.
The term “perhaloalkyl” as used herein, alone or in combination, refers to an alkyl group where all of the hydrogen atoms are replaced by halogen atoms.
The terms “sulfonate,” “sulfonic acid,” and “sulfonic,” as used herein, alone or in combination, refer the —SO3H group and its anion as the sulfonic acid is used in salt formation.
The term “sulfanyl,” as used herein, alone or in combination, refers to —S—.
The term “sulfinyl,” as used herein, alone or in combination, refers to —S(O)—.
The term “sulfonyl,” as used herein, alone or in combination, refers to —S(O)2—.
The term “N-sulfonamido” refers to a RS(═O)2NR′— group with R and R′ as defined herein.
The term “S-sulfonamido” refers to a —S(═O)2NRR′, group, with R and
R′ as defined herein.
The terms “thia” and “thio,” as used herein, alone or in combination, refer to a —S— group or an ether wherein the oxygen is replaced with sulfur. The oxidized derivatives of the thio group, namely sulfinyl and sulfonyl, are included in the definition of thia and thio.
The term “thiol,” as used herein, alone or in combination, refers to an —SH group.
The term “thiocarbonyl,” as used herein, when alone includes thioformyl —C(S)H and in combination is a —C(S)— group.
The term “N-thiocarbamyl” refers to an ROC(S)NR′— group, with R and R′ as defined herein.
The term “O-thiocarbamyl” refers to a —OC(S)NRR′, group with R and R′ as defined herein.
The term “thiocyanato” refers to a —CNS group.
The term “trihalomethanesulfonamido” refers to a X3CS(O)2NR— group with X is a halogen and R as defined herein.
The term “trihalomethanesulfonyl” refers to a X3CS(O)2— group where X is a halogen.
The term “trihalomethoxy” refers to a X3CO— group where X is a halogen.
The term “trisubstituted silyl,” as used herein, alone or in combination, refers to a silicone group substituted at its three free valences with groups as listed herein under the definition of substituted amino. Examples include trimethysilyl, tert-butyldimethylsilyl, triphenylsilyl and the like.
Any definition herein may be used in combination with any other definition to describe a composite structural group. By convention, the trailing element of any such definition is that which attaches to the parent moiety. For example, the composite group alkylamido would represent an alkyl group attached to the parent molecule through an amido group, and the term alkoxyalkyl would represent an alkoxy group attached to the parent molecule through an alkyl group.
When a group is defined to be “null,” what is meant is that said group is absent.
The term “optionally substituted” means the anteceding group may be substituted or unsubstituted. When substituted, the substituents of an “optionally substituted” group may include, without limitation, one or more substituents independently selected from the following groups or a particular designated set of groups, alone or in combination: lower alkyl, lower alkenyl, lower alkynyl, lower alkanoyl, lower heteroalkyl, lower heterocycloalkyl, lower haloalkyl, lower haloalkenyl, lower haloalkynyl, lower perhaloalkyl, lower perhaloalkoxy, lower cycloalkyl, phenyl, aryl, aryloxy, lower alkoxy, lower haloalkoxy, oxo, lower acyloxy, carbonyl, carboxyl, lower alkylcarbonyl, lower carboxyester, lower carboxamido, cyano, hydrogen, halogen, hydroxy, amino, lower alkylamino, arylamino, amido, nitro, thiol, lower alkylthio, lower haloalkylthio, lower perhaloalkylthio, arylthio, sulfonate, sulfonic acid, trisubstituted silyl, N3, SH, SCH3, C(O)CH3, CO2CH3, CO2H, pyridinyl, thiophene, furanyl, lower carbamate, and lower urea. Two substituents may be joined together to form a fused five-, six-, or seven-membered carbocyclic or heterocyclic ring consisting of zero to three heteroatoms, for example forming methylenedioxy or ethylenedioxy. An optionally substituted group may be unsubstituted (e.g., —CH2CH3), fully substituted (e.g., —CF2CF3), monosubstituted (e.g., —CH2CH2F) or substituted at a level anywhere in-between fully substituted and monosubstituted (e.g., —CH2CF3). Where substituents are recited without qualification as to substitution, both substituted and unsubstituted forms are encompassed. Where a substituent is qualified as “substituted,” the substituted form is specifically intended. Additionally, different sets of optional substituents to a particular moiety may be defined as needed; in these cases, the optional substitution will be as defined, often immediately following the phrase, “optionally substituted with.”
The term R or the term R′, appearing by itself and without a number designation, unless otherwise defined, refers to a moiety selected from the group consisting of hydrogen, alkyl, cycloalkyl, heteroalkyl, aryl, heteroaryl and heterocycloalkyl, any of which may be optionally substituted. Such R and R′ groups should be understood to be optionally substituted as defined herein. Whether an R group has a number designation or not, every R group, including R, R′ and Rn where n=(1, 2, 3, . . . n), every substituent, and every term should be understood to be independent of every other in terms of selection from a group. Should any variable, substituent, or term (e.g. aryl, heterocycle, R, etc.) occur more than one time in a formula or generic structure, its definition at each occurrence is independent of the definition at every other occurrence. Those of skill in the art will further recognize that certain groups may be attached to a parent molecule or may occupy a position in a chain of elements from either end as written. Thus, by way of example only, an unsymmetrical group such as —C(O)N(R)— may be attached to the parent moiety at either the carbon or the nitrogen.
Asymmetric centers exist in the compounds disclosed herein. These centers are designated by the symbols “R” or “S,” depending on the configuration of substituents around the chiral carbon atom. It should be understood that the invention encompasses all stereochemical isomeric forms, including diastereomeric, enantiomeric, and epimeric forms, as well as d-isomers and 1-isomers, and mixtures thereof. Individual stereoisomers of compounds can be prepared synthetically from commercially available starting materials which contain chiral centers or by preparation of mixtures of enantiomeric products followed by separation such as conversion to a mixture of diastereomers followed by separation or recrystallization, chromatographic techniques, direct separation of enantiomers on chiral chromatographic columns, or any other appropriate method known in the art. Starting compounds of particular stereochemistry are either commercially available or can be made and resolved by techniques known in the art. Additionally, the compounds disclosed herein may exist as geometric isomers. The present invention includes all cis, trans, syn, anti, entgegen (E), and zusammen (Z) isomers as well as the appropriate mixtures thereof. Additionally, compounds may exist as tautomers; all tautomeric isomers are provided by this invention. Additionally, the compounds disclosed herein can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms.
The term “bond” refers to a covalent linkage between two atoms, or two moieties when the atoms joined by the bond are considered to be part of larger substructure. A bond may be single, double, or triple unless otherwise specified. A dashed line between two atoms in a drawing of a molecule indicates that an additional bond may be present or absent at that position.
The term “disease” as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disorder” and “condition” (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms, and causes the human or animal to have a reduced duration or quality of life.
The term “NF-κB-mediated disease,” refers to a disease in which NF-κB plays an active role in the disease pathology. NF-κB-mediated diseases include diseases in which multiple biological pathways and/or processes in addition to NF-κB-mediated processes contribute to the disease pathology. A NF-κB-mediated disease may be completely or partially mediated by modulating the activity or amount of NF-κB. In particular, a NF-κB-mediated disease is one in which modulation of NF-κB results in some effect on the underlying disease e.g., administration of a NF-κB modulator results in some improvement in at least some of the patients being treated. The term “NF-κB-mediated disease” also refers to the following diseases, even though the compounds disclosed herein exert their effects through biological pathways and/or processes other than NF-κB: muscular dystrophy, arthritis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type 2 diabetes, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, muscle wasting, catabolic disorders, obesity, fetal growth retardation, hypercholesterolemia, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, asthma, chronic obstructive pulmonary disease, Sjogren's syndrome, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, gut diseases, peritoneal endometriosis, skin diseases, nasal sinusitis, mesothelioma, anhidrotic ecodermal dysplasia-ID, behcet's disease, incontinentia pigmenti, tuberculosis, asthma, crohn's disease, colitis, ocular allergy, appendicitis, paget's disease, pancreatitis, periodonitis, endometriosis, inflammatory bowel disease, inflammatory lung disease, silica-induced diseases, sleep apnea, AIDS, HIV-1, autoimmune diseases, antiphospholipid syndrome, lupus, lupus nephritis, familial mediterranean fever, hereditary periodic fever syndrome, psychosocial stress diseases, neuropathological diseases, familial amyloidotic polyneuropathy, inflammatory neuropathy, parkinson's disease, multiple sclerosis, alzheimer's disease, amyotropic lateral sclerosis, huntington's disease, cataracts, and hearing loss.
The term “combination therapy” means the administration of two or more therapeutic agents to treat a therapeutic condition or disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.
“NF-κB modulator is used herein to refer to a compound that exhibits an EC50 with respect to NF-κB activity of no more than about 100 μM and more typically not more than about 50 μM, as measured in the NF-κB inhibitor assays described generally hereinbelow. “EC50” is that concentration of modulator which either activates or reduces the activity or increases or decreases the amount of an enzyme (e.g., (NF-κB)) to half-maximal level. Certain compounds disclosed herein have been discovered to exhibit modulatory activity against NF-κB. In certain embodiments, compounds will exhibit an EC50 with respect to NF-κB of no more than about 10 μM; in further embodiments, compounds will exhibit an EC50 with respect to NF-κB of no more than about 5 μM; in yet further embodiments, compounds will exhibit an EC50 with respect to NF-κB of not more than about 1 μM; in yet further embodiments, compounds will exhibit an EC50 with respect to NF-κB of not more than about 200 nM, as measured in the NF-κB assay described herein.
The phrase “therapeutically effective” is intended to qualify the amount of active ingredients used in the treatment of a disease or disorder. This amount will achieve the goal of reducing or eliminating the said disease or disorder.
The term “therapeutically acceptable” refers to those compounds (or salts, prodrugs, tautomers, zwitterionic forms, etc.) which are suitable for use in contact with the tissues of patients without undue toxicity, irritation, and allergic response, are commensurate with a reasonable benefit/risk ratio, and are effective for their intended use.
As used herein, reference to “treatment” of a patient is intended to include prophylaxis. The term “patient” means all mammals including humans. Examples of patients include humans, cows, dogs, cats, goats, sheep, pigs, and rabbits. Preferably, the patient is a human.
The term “prodrug” refers to a compound that is made more active in vivo. Certain compounds disclosed herein may also exist as prodrugs, as described in Hydrolysis in Drug and Prodrug Metabolism: Chemistry, Biochemistry, and Enzymology (Testa, Bernard and Mayer, Joachim M. Wiley-VHCA, Zurich, Switzerland 2003). Prodrugs of the compounds described herein are structurally modified forms of the compound that readily undergo chemical changes under physiological conditions to provide the compound. Additionally, prodrugs can be converted to the compound by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to a compound when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent. Prodrugs are often useful because, in some situations, they may be easier to administer than the compound, or parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not. The prodrug may also have improved solubility in pharmaceutical compositions over the parent drug. A wide variety of prodrug derivatives are known in the art, such as those that rely on hydrolytic cleavage or oxidative activation of the prodrug. An example, without limitation, of a prodrug would be a compound which is administered as an ester (the “prodrug”), but then is metabolically hydrolyzed to the carboxylic acid, the active entity. Additional examples include peptidyl derivatives of a compound.
The compounds disclosed herein can exist as therapeutically acceptable salts. The present invention includes compounds listed above in the form of salts, including acid addition salts. Suitable salts include those formed with both organic and inorganic acids. Such acid addition salts will normally be pharmaceutically acceptable. However, salts of non-pharmaceutically acceptable salts may be of utility in the preparation and purification of the compound in question. Basic addition salts may also be formed and be pharmaceutically acceptable. For a more complete discussion of the preparation and selection of salts, refer to Pharmaceutical Salts Properties, Selection, and Use (Stahl, P. Heinrich. Wiley-VCHA, Zurich, Switzerland, 2002).
The terms “therapeutically acceptable salt,” or “salt,” as used herein, represents salts or zwitterionic forms of the compounds disclosed herein which are water or oil-soluble or dispersible and therapeutically acceptable as defined herein. The salts can be prepared during the final isolation and purification of the compounds or separately by reacting the appropriate compound in the form of the free base with a suitable acid. Representative acid addition salts include acetate, adipate, alginate, L-ascorbate, aspartate, benzoate, benzenesulfonate (besylate), bisulfate, butyrate, camphorate, camphorsulfonate, citrate, digluconate, formate, fumarate, gentisate, glutarate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethansulfonate (isethionate), lactate, maleate, malonate, DL-mandelate, mesitylenesulfonate, methanesulfonate, naphthylenesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylproprionate, phosphonate, picrate, pivalate, propionate, pyroglutamate, succinate, sulfonate, tartrate, L-tartrate, trichloroacetate, trifluoroacetate, phosphate, glutamate, bicarbonate, para-toluenesulfonate (p-tosylate), and undecanoate. Also, basic groups in the compounds disclosed herein can be quaternized with methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides; dimethyl, diethyl, dibutyl, and diamyl sulfates; decyl, lauryl, myristyl, and steryl chlorides, bromides, and iodides; and benzyl and phenethyl bromides. Examples of acids which can be employed to form therapeutically acceptable addition salts include inorganic acids such as hydrochloric, hydrobromic, sulfuric, and phosphoric, and organic acids such as oxalic, maleic, succinic, and citric. Salts can also be formed by coordination of the compounds with an alkali metal or alkaline earth ion. Hence, the present invention contemplates sodium, potassium, magnesium, and calcium salts of the compounds disclosed herein, and the like.
Basic addition salts can be prepared during the final isolation and purification of the compounds by reacting a carboxy group with a suitable base such as the hydroxide, carbonate, or bicarbonate of a metal cation or with ammonia or an organic primary, secondary, or tertiary amine. The cations of therapeutically acceptable salts include lithium, sodium, potassium, calcium, magnesium, and aluminum, as well as nontoxic quaternary amine cations such as ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, diethylamine, ethylamine, tributylamine, pyridine, N,N-dimethylaniline, N-methylpiperidine, N-methylmorpholine, dicyclohexylamine, procaine, dibenzylamine, N,N-dibenzylphenethylamine, 1-ephenamine, and N,N′-dibenzylethylenediamine. Other representative organic amines useful for the formation of base addition salts include ethylenediamine, ethanolamine, diethanolamine, piperidine, and piperazine.
In certain embodiments, the salts may include hydrochloride, hydrobromide, sulfonate, citrate, tartrate, phosphonate, lactate, pyruvate, acetate, succinate, oxalate, fumarate, malate, oxaloacetate, methanesulfonate, ethanesulfonate, p-toluenesulfonate, benzenesulfonate and isethionate salts of compounds disclosed herein. A salt of a compound can be made by reacting the appropriate compound in the form of the free base with the appropriate acid.
While it may be possible for the compounds of the subject invention to be administered as the raw chemical, it is also possible to present them as a pharmaceutical formulation. Accordingly, provided herein are pharmaceutical formulations which comprise one or more of certain compounds disclosed herein, or one or more pharmaceutically acceptable salts, esters, prodrugs, amides, or solvates thereof, together with one or more pharmaceutically acceptable carriers thereof and optionally one or more other therapeutic ingredients. The carrier(s) must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Proper formulation is dependent upon the route of administration chosen. Any of the well-known techniques, carriers, and excipients may be used as suitable and as understood in the art; e.g., in Remington's Pharmaceutical Sciences. The pharmaceutical compositions disclosed herein may be manufactured in any manner known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
The formulations include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous, intraarticular, and intramedullary), intraperitoneal, transmucosal, transdermal, rectal and topical (including dermal, buccal, sublingual and intraocular) administration although the most suitable route may depend upon for example the condition and disorder of the recipient. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Typically, these methods include the step of bringing into association a compound of the subject invention or a pharmaceutically acceptable salt, ester, amide, prodrug or solvate thereof (“active ingredient”) with the carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
Formulations of the compounds disclosed herein suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.
Pharmaceutical preparations which can be used orally include tablets, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. Tablets may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with binders, inert diluents, or lubricating, surface active or dispersing agents. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein. All formulations for oral administration should be in dosages suitable for such administration. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
The compounds may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in powder form or in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or sterile pyrogen-free water, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
Formulations for parenteral administration include aqueous and non-aqueous (oily) sterile injection solutions of the active compounds which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
For oral or parenteral use, the compounds may be formulated as nanoparticle preparations. Such nanoparticle preparations can include, for example, nanoshere encapsulations of active compounds, inactive nanoparticles to which active compounds can be tethered, or nanoscale powders of active compounds. Nanoparticle preparations can be used to increase the bioavailability of the active compounds, control the rate of release of the active compounds, or deliver active compounds to a particular location in the body. See A. Dove, “An Easy Pill to Swallow”, Drug Discovery & Development Magazine: 11(11), November, 2008, pp. 22-24.
In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection. Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
For buccal or sublingual administration, the compositions may take the form of tablets, lozenges, pastilles, or gels formulated in conventional manner. Such compositions may comprise the active ingredient in a flavored basis such as sucrose and acacia or tragacanth.
The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter, polyethylene glycol, or other glycerides.
Certain compounds disclosed herein may be administered topically, that is by non-systemic administration. This includes the application of a compound disclosed herein externally to the epidermis or the buccal cavity and the instillation of such a compound into the ear, eye and nose, such that the compound does not significantly enter the blood stream. In contrast, systemic administration refers to oral, intravenous, intraperitoneal and intramuscular administration.
Formulations suitable for topical administration include liquid or semi-liquid preparations suitable for penetration through the skin to the site of inflammation such as gels, liniments, lotions, creams, ointments or pastes, and drops suitable for administration to the eye, ear or nose. The active ingredient for topical administration may comprise, for example, from 0.001% to 10% w/w (by weight) of the formulation. In certain embodiments, the active ingredient may comprise as much as 10% w/w. In other embodiments, it may comprise less than 5% w/w. In certain embodiments, the active ingredient may comprise from 2% w/w to 5% w/w. In other embodiments, it may comprise from 0.1% to 1% w/w of the formulation.
Formulations for topical administration in the mouth, for example buccally or sublingually, include lozenges comprising the active ingredient in a flavored basis such as sucrose and acacia or tragacanth, and pastilles comprising the active ingredient in a basis such as gelatin and glycerin or sucrose and acacia.
For administration by inhalation, compounds may be conveniently delivered from an insufflator, nebulizer pressurized packs or other convenient means of delivering an aerosol spray. Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Alternatively, for administration by inhalation or insufflation, the compounds according to the invention may take the form of a dry powder composition, for example a powder mix of the compound and a suitable powder base such as lactose or starch. The powder composition may be presented in unit dosage form, in for example, capsules, cartridges, gelatin or blister packs from which the powder may be administered with the aid of an inhalator or insufflator.
Preferred unit dosage formulations are those containing an effective dose, as herein below recited, or an appropriate fraction thereof, of the active ingredient.
It should be understood that in addition to the ingredients particularly mentioned above, the formulations described above may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
Compounds may be administered orally or via injection at a dose of from 0.1 to 500 mg/kg per day. The dose range for adult humans is generally from 5 mg to 2 g/day. Tablets or other forms of presentation provided in discrete units may conveniently contain an amount of one or more compounds which is effective at such dosage or as a multiple of the same, for instance, units containing 5 mg to 500 mg, usually around 10 mg to 200 mg.
The amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
The compounds can be administered in various modes, e.g. orally, topically, or by injection. The precise amount of compound administered to a patient will be the responsibility of the attendant physician. The specific dose level for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diets, time of administration, route of administration, rate of excretion, drug combination, the precise disorder being treated, and the severity of the indication or condition being treated. Also, the route of administration may vary depending on the condition and its severity.
In certain instances, it may be appropriate to administer at least one of the compounds described herein (or a pharmaceutically acceptable salt, ester, or prodrug thereof) in combination with another therapeutic agent. By way of example only, if one of the side effects experienced by a patient upon receiving one of the compounds herein is hypertension, then it may be appropriate to administer an anti-hypertensive agent in combination with the initial therapeutic agent. Or, by way of example only, the therapeutic effectiveness of one of the compounds described herein may be enhanced by administration of an adjuvant (i.e., by itself the adjuvant may only have minimal therapeutic benefit, but in combination with another therapeutic agent, the overall therapeutic benefit to the patient is enhanced). Or, by way of example only, the benefit of experienced by a patient may be increased by administering one of the compounds described herein with another therapeutic agent (which also includes a therapeutic regimen) that also has therapeutic benefit. By way of example only, in a treatment for diabetes involving administration of one of the compounds described herein, increased therapeutic benefit may result by also providing the patient with another therapeutic agent for diabetes. In any case, regardless of the disease, disorder or condition being treated, the overall benefit experienced by the patient may simply be additive of the two therapeutic agents or the patient may experience a synergistic benefit.
In any case, the multiple therapeutic agents (at least one of which is a compound disclosed herein) may be administered in any order or even simultaneously. If simultaneously, the multiple therapeutic agents may be provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills). One of the therapeutic agents may be given in multiple doses, or both may be given as multiple doses. If not simultaneous, the timing between the multiple doses may be any duration of time ranging from a few minutes to four weeks.
Thus, in another aspect, certain embodiments provide methods for treating NF-κB-mediated disorders in a human or animal subject in need of such treatment comprising administering to said subject an amount of a compound disclosed herein effective to reduce or prevent said disorder in the subject, in combination with at least one additional agent for the treatment of said disorder that is known in the art. In a related aspect, certain embodiments provide therapeutic compositions comprising at least one compound disclosed herein in combination with one or more additional agents for the treatment of NF-κB-mediated disorders.
Specific diseases to be treated by the compounds, compositions, and methods disclosed herein include ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, muscular dystrophy, muscle wasting, catabolic disorders, type 1 diabetes, type 2 diabetes, obesity, fetal growth retardation, hypercholesterolemia, atherosclerosis, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, chronic obstructive pulmonary disease, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, leptospiriosis renal disease, gut diseases, peritoneal endometriosis, skin diseases, nasal sinusitis, mesothelioma, anhidrotic ecodermal dysplasia-ID, behcet's disease, incontinentia pigmenti, tuberculosis, asthma, arthritis, crohn's disease, colitis, ocular allergy, glaucoma, appendicitis, paget's disease, pancreatitis, periodonitis, endometriosis, inflammatory bowel disease, inflammatory lung disease, sepsis, silica-induced diseases, sleep apnea, AIDS, HIV-1, autoimmune diseases, antiphospholipid syndrome, lupus, lupus nephritis, familial mediterranean fever, hereditary periodic fever syndrome, psychosocial stress diseases, neuropathological diseases, familial amyloidotic polyneuropathy, inflammatory neuropathy, traumatic brain injury, spinal cord injury, parkinson's disease, multiple sclerosis, rheumatic disease, alzheimer's disease, amyotropic lateral sclerosis, huntington's disease, retinal disease, cataracts, hearing loss, and cancer.
Besides being useful for human treatment, certain compounds and formulations disclosed herein may also be useful for veterinary treatment of companion animals, exotic animals and farm animals, including mammals, rodents, and the like. More preferred animals include horses, dogs, and cats.
All references, patents or applications, U.S. or foreign, cited in the application are hereby incorporated by reference as if written herein in their entireties. Where any inconsistencies arise, material literally disclosed herein controls.
General Synthetic Methods for Preparing Compounds
The following schemes can be used to practice the present invention.
Figure US09198921-20151201-C00017
Examples 6-7, 15, 20, and 33-34 can be synthesized according to scheme I.
Figure US09198921-20151201-C00018
Examples 8, 16-17, 26, and 35-36 can be synthesized according to scheme II.
Figure US09198921-20151201-C00019
Examples 9, 14, 19, 27, and 37-38 can be synthesized according to scheme III.
Figure US09198921-20151201-C00020
Examples 11, 13, 18, 28, and 39-40 can be synthesized according to scheme IV.
Figure US09198921-20151201-C00021
Example 24 can be synthesized according to scheme V.
Figure US09198921-20151201-C00022
Examples 1-3, 25, and 29-32 can be synthesized according to scheme VI.
Figure US09198921-20151201-C00023
Preparation 1 can be synthesized according to scheme VII.
Figure US09198921-20151201-C00024
Example 10 can be synthesized according to scheme VIII.
The invention is further illustrated by the following examples. All IUPAC names were generated using CambridgeSoft's ChemDraw 10.0.
Preparation 1 2-oxo-2-((6S,10R,13S)-6,10,13-trimethyl-3-oxo-6,7,8,10,12,13,14,15-octahydro-3H-cyclopenta[a]phenanthren-17-yl)ethyl acetate
Figure US09198921-20151201-C00025

Step 1
Figure US09198921-20151201-C00026
2-((6S,10R,13S)-17-hydroxy-6,10,13-trimethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate: (see Tetrahedron Letters, 2001, 42 (14): 2639-2642). Alternatively, methyl prednisolone 21-acetate is dissolved in a mixture of dimethylformamide and tetrahydrofuran and cooled in an ice bath. SO2 is bubbled into methanesulfonyl chloride and the mixture is added dropwise to the solution containing the solution of methyl prednisolone 21-acetate. The title product can then be isolated by standard aqueous workup.
Step 2
Figure US09198921-20151201-C00027
(2′R,4′R,6S,10R,13S)-2′-acetyl-2′,6,10,13-tetramethyl-7,8,10,12,13,14,15,16-octahydrospiro[cyclopenta[a]phenanthrene-17,4′-[1,3]dioxane]-3,5′(6H)-dione: 2-((6S,10R,13S)-17-hydroxy-6,10,13-trimethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate is dissolved in toluene and heated with 1.5 equivalents of ethyl orthoacetate and a trace of pyridinium hydrochloride. Ethanol is distilled off the reaction mixture to drive it to completion.
Step 3
Figure US09198921-20151201-C00028
(6S,10R,13S)-17-(2-hydroxyacetyl)-6,10,13-trimethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl acetate: The reaction mixture from step 3 is concentrated, dissolved in tetrahydrofuran, and treated with dilute hydrochloric acid. Standard aqueous workup yields the title compound.
Step 4
Figure US09198921-20151201-C00029
2-oxo-2-((6S,10R,13S)-6,10,13-trimethyl-3-oxo-6,7,8,10,12,13,14,15-octahydro-3H-cyclopenta[a]phenanthren-17-yl)ethyl acetate: (6S,10R,13S)-17-(2-hydroxyacetyl)-6,10,13-trimethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl acetate is heated with 2 equivalents of potassium carbonate in dimethyl formamide. Standard aqueous workup yields the title compound.
EXAMPLE 1 (10S,13S,17R)-17-hydroxy-17-(2-hydroxyacetyl)-10,13-dimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one
Figure US09198921-20151201-C00030

Step 1
Figure US09198921-20151201-C00031
(10S,13S,17R)-17-hydroxy-17-(2-hydroxyacetyl)-10,13-dimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one: Commercially available as Anecortave acetate. The title compound can be synthesized according to the procedure of Example 8, Step 2, substituting 2-((10S,13S,17R)-17-hydroxy-10,13-dimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate for 2-oxo-2-((10S,13S,16R,17S)-10,13,16-trimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)ethyl acetate.
EXAMPLE 2 (10S,13S,17R)-17-hydroxy-17-(2-hydroxyacetyl)-10,13-dimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one
Figure US09198921-20151201-C00032
(10S,13S,17R)-17-hydroxy-17-(2-hydroxyacetyl)-10,13-dimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one: The title compound can be synthesized according to the procedures of Example 3, Step 1 and Example 1, Step 1, substituting prednisolone acetate for hydrocortisone acetate.
EXAMPLE 3 2-(10S,13S,17R)-17-hydroxy-10,13-dimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate
Figure US09198921-20151201-C00033

Step 1
Figure US09198921-20151201-C00034
2-((10S,13S,17R)-17-hydroxy-10,13-dimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate: The title compound can be synthesized from hydrocortisone acetate according to the procedure disclosed in EP 0097328.405 g (1 mol) of hydrocortisone acetate is added to a mixture of 2 liters of N,N-dimethylformamide and 350 ml of pyridine, and with stiffing at room temperature, 260 g of methanesulfonyl chloride is added. The reaction mixture is heated, maintained at 80 to 85° C. for 1 hour, and then cooled to room temperature. Methanol (7 liters) is added. The precipitated crystals are separated by filtration, washed with methanol and water, and dried under reduced pressure to give the title compound.
EXAMPLE 6 (10S,13S,16R,17S)-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one
Figure US09198921-20151201-C00035

Step 1
Figure US09198921-20151201-C00036
(10S,13S,16R,17S)-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one: A solution of the product from Example 7, step 2 in methylene chloride and methanol (1:3 methylene chloride/methanol) is stirred under an inert atmosphere and cooled in an ice bath. Aqueous potassium carbonate is added by syringe. The reaction is stirred at 5° C. for 2 hours. The reaction is then neutralized with 1N HCl and concentrated. After partitioning between water and methylene chloride, the product solution is dried over anhydrous magnesium sulfate, filtered and evaporated to give the title compound.
EXAMPLE 7 2-oxo-2-((10S,13S,16R)-10,13,16-trimethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl)ethyl acetate
Figure US09198921-20151201-C00037

Step 1
Figure US09198921-20151201-C00038
2-((10S,13S,16R)-10,13,16-trimethyl-3-oxo-7,8,12,13,15,16-hexahydro-3H-cyclopenta[a]phenanthren-17(6H,10H,14H)-ylidene)-2-(trimethylsilyloxy)ethyl acetate: (see K. P. Shephard, U.S. Pat. No. 4,975,536; Dec. 4, 1990; Preparation 1, col. 8) Into predried reactor 1 was added 36.64 grams (100 mmole) of 2-((10S,13S)-10,13-dimethyl-3-oxo-6,7,8,10,12,13,14,15-octahydro-3H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate (commercial product from Pfizer). The starting material was dissolved in 200 ml of anhydrous tetrahydrofuran and 200 ml of anhydrous dichloromethane. Trimethylsilyl imidazole, (20.0 ml, 136 mmole), was added. This solution was cooled to −50° C. under a small nitrogen flow.
Into predried reactor 2 was added copper II propionate (2.10 grams, 10.0 mmole), 150 ml of anhydrous tetrahydrofuran, and anhydrous 1,3-dimethyl-3,4,5,6-tetrahydro-2(1H)-pyrimidinone. The mixture was cooled to −50° C. and methyl magnesium chloride (3M, 10.0 ml) was added dropwise over approximately 5 minutes. The mixture was stirred for approximately 10 minutes. The contents of reactor 2 were transferred to reactor 1 via cannula quickly (approximately 30 sec.), and reactor 2 was rinsed with 10 ml of anhydrous tetrahydrofuran and this was also cannulated into reactor 1. A pump was set up with methyl magnesium chloride (3M, 45.0 ml) and pumped into reactor 1 over 45 min (pump setting at 1.0 ml/min). Reactor 1 was stirred further at −50° C. for 1 hour, then warmed to −30° C. overnight.
Toluene (1 L) was added and the temperature brought to 0° C. The mixture was extracted with 2×500 ml of 5% acetic acid (cold), then with 200 ml of 25% sodium chloride. The aqueous phases were back extracted with 300 ml of toluene. The combined toluene extracts were dried over magnesium sulfate, filtered, and concentrated to a viscous oil. Yield—57.8 grams.
Step 2
Figure US09198921-20151201-C00039
2-oxo-2-((10S,13S,16R,17S)-10,13,16-trimethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl)ethyl acetate: The crude product from step 1 is dissolved in ethyl acetate, and slurried with aqueous 1N HCl until hydrolysis is complete. The aqueous acid is neutralized with aqueous potassium bicarbonate, and the ethyl acetate phase is dried, filtered, and concentrated to a semi-solid.
EXAMPLE 8 (10S,13S,16R,17S)-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one
Figure US09198921-20151201-C00040

Step 1
Figure US09198921-20151201-C00041
2-oxo-2-((10S,13S,16R,17S)-10,13,16-trimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)ethyl acetate: A mixture of 3.3 g (8.6 mM) of 2-oxo-2-((10S,13S,16R,17S)-10,13,16-trimethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl)ethyl acetate, chlorotris(triphenylphosphine)rhodium(I) (Wilkinson's Catalyst, 480 mg, 0.52 mM), triethylsilane (1.4 mL, 1.0 g, 8.8 mM) and methylene chloride (15 mL) was warmed to 40° C. and stirred until most of the starting material was gone, as determined by thin layer chromatography. The reaction was evaporated in vacuo and chromatographed on fine silica gel (600 g) in 10-15% ethyl acetate in methylene chloride. One fraction of 700 mL was collected, followed by twelve 200 mL fractions. A 1.0 g quantity of desired product (30% yield) was obtained by evaporation of fractions 6-12. (Starting material was obtained from fraction 13, 0.7 g, 20% recovery). NMR (500 MHz, CDCl3, TMS): δ 0.68 (s, 3H), 0.98 (d, 3H, J=6.5 Hz), 1.12 (m, 1H), 1.33 (s, 3H), 1.47 (m, 1H), 1.57 (m, 1H), 1.69 (m, 1H), 1.99 (m, 1H), 2.18 (s, 3H), 2.07-2.29 (m, 6H), 2.36 (d, 1H), 2.50 (m, 3H), 2.79 (m, 1H), 4.48 (d, 1H, J=17 Hz), 4.73 (d, 1H, J=17 Hz), 5.50 (s, 1H), 5.75 (s, 1H).
Step 2
Figure US09198921-20151201-C00042
(10S,13S,16R,17S)-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one: A solution of 2-oxo-2-((10S,13 S,16R,17S)-10,13,16-trimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)ethyl acetate (1.0 g, 2.6 mM) in methylene chloride (5 mL) and methanol (15 mL) was put under an inert atmosphere and cooled in an ice bath. 1 mL of 1 M aqueous potassium carbonate was added by syringe. The reaction was stirred at 5° C. for 2 h. The reaction was then neutralized with 1N HCl and concentrated. After partitioning between water and methylene chloride, the product solution was dried over anhydrous magnesium sulfate, filtered and evaporated. Crystallization from ethyl acetate yielded a first crop of 0.33 g product. NMR (500 MHz, CDCl3, TMS): δ 0.67 (s, 3H), 1.01 (d, 3H, J=7 Hz), 1.13 (m, 1H), 1.33 (s, 3H), 1.47-1.80 (m, 3, H), 2.00 (m, 1H), 2.06-2.24 (m, 6H), 2.37 (d, 1H), 2.45-2.60 (m, 3H), 2.82 (m, 1H), 3.30 (m, 1H), 4.20 (m, 2H), 5.50 (d, 1H, J=5 Hz), 5.76 (s, 1H).
EXAMPLE 9 (10S,13S,16R,17R)-17-hydroxy-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one
Figure US09198921-20151201-C00043

Step 1
Figure US09198921-20151201-C00044
2-((10S,13S,16R,17R)-17-hydroxy-10,13,16-trimethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate: (Z)-2-((10S,13S,16R)-10,13,16-trimethyl-3-oxo-7,8,12,13,15,16-hexahydro-3H-cyclopenta[a]phenanthren-17(6H,10H,14H)-ylidene)-2-(trimethylsilyloxy)ethyl acetate is dissolved in methylene chloride and the mixture is cooled to zero degrees Celsius. A solution of m-chloroperbenzoic acid in methylene chloride is added dropwise and the mixture is stirred for 4 hours. The organic phase was washed with aqueous acetic acid and then aqueous bisulfite. The organic phase was concentrated and chromatographed on silica gel to yield the title compound.
Step 2
Figure US09198921-20151201-C00045
(10S,13S,16R,17R)-17-hydroxy-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one: Prepared according to Example 8, Step 2 substituting 2-((10S,13S,16R,17R)-17-hydroxy-10,13,16-trimethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate for 2-oxo-2-((10S,13S,16R,17S)-10,13,16-trimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)ethyl acetate.
EXAMPLE 10 (10S,13S,16S,17R)-17-hydroxy-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one
Figure US09198921-20151201-C00046
EXAMPLE 11 (10S,13S,16R,17R)-17-hydroxy-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one
Figure US09198921-20151201-C00047
EXAMPLE 12 (10S,13S,16S,17R)-17-hydroxy-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one
Figure US09198921-20151201-C00048
EXAMPLE 13 (10S,13S,16R,17R)-17-hydroxy-17-(2-hydroxyacetyl)-10,13-dimethyl-16-propyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one
Figure US09198921-20151201-C00049
EXAMPLE 14 (10S,13S,16R,17R)-17-hydroxy-17-(2-hydroxyacetyl)-10,13-dimethyl-16-propyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one
Figure US09198921-20151201-C00050
EXAMPLE 15 (10S,13S,16R,17S)-17-(2-hydroxyacetyl)-10,13-dimethyl-16-propyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one
Figure US09198921-20151201-C00051
EXAMPLE 16 (10S,13S,16R,17S)-17-(2-hydroxyacetyl)-10,13-dimethyl-16-propyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one
Figure US09198921-20151201-C00052
EXAMPLE 17 (10S,13S,16R,17S)-17-(2-hydroxyacetyl)-10,13-dimethyl-16-phenyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one
Figure US09198921-20151201-C00053
EXAMPLE 18 (10S,13S,16S,17R)-17-hydroxy-17-(2-hydroxyacetyl)-10,13-dimethyl-16-phenyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one
Figure US09198921-20151201-C00054
EXAMPLE 19 (10S,13S,16S,17R)-17-hydroxy-17-(2-hydroxyacetyl)-10,13-dimethyl-16-phenyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one
Figure US09198921-20151201-C00055
EXAMPLE 20 (10S,13S,16R,17S)-17-(2-hydroxyacetyl)-10,13-dimethyl-16-phenyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one
Figure US09198921-20151201-C00056
EXAMPLE 21 (10S,13S,16S,17S)-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one
Figure US09198921-20151201-C00057
EXAMPLE 22 (10S,13S,16S,17S)-17-(2-hydroxyacetyl)-10,13,16-trimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one
Figure US09198921-20151201-C00058
EXAMPLE 23 2-oxo-2-((10S,13S,16S,17S)-10,13,16-trimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)ethyl acetate
Figure US09198921-20151201-C00059
EXAMPLE 24 (10S,13S,16R,17S)-17-(2-hydroxyacetyl)-10,13,16,17-tetramethyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one
Figure US09198921-20151201-C00060

Step 1
Figure US09198921-20151201-C00061
2-oxo-2-((10S,13S,16R,17S)-10,13,16,17-tetramethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)ethyl acetate: A mixture of 2-((10S,13S)-10,13-dimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15-decahydro-1H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate (150 g) and copper propionate (1.9M in THF (90 ml) is cooled in an ice acetone bath. Methyl magnesium chloride (1.96M in THF, 240 ml) is added dropwise for 30 min. After 1 hour, the reaction is quenched with methyl iodide (100 g) in 200 ml THF. The reaction mixture is then partitioned with water and toluene. The separated organic phase is washed with water, dried over sodium sulfate and concentrated. The residue is crystallized from ether and hexane to give the title compound.
Step 2
Figure US09198921-20151201-C00062
(10S,13S,16R,17S)-17-(2-hydroxyacetyl)-10,13,16,17-tetramethyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one: 2-oxo-2-((10S,13S,16R,17S)-10,13,16,17-tetramethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)ethyl acetate (144 g) is stirred in 1500 ml methanol and treated with sodium methoxide (25%, 5 ml) for 30 minutes. The mixture is partitioned between methylene chloride and sodium bicarbonate. The organic phase is separated and washed with sodium bicarbonate, dried over sodium sulfate, and concentrated to give the title compound.
EXAMPLE 25 2-(10S,13S,17R)-17-hydroxy-10,13-dimethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate
Figure US09198921-20151201-C00063
EXAMPLE 26 2-oxo-2-((10S,13S,16R,17S)-10,13,16-trimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)ethyl acetate
Figure US09198921-20151201-C00064
EXAMPLE 27 2-((10S,13S,16R,17R)-17-hydroxy-10,13,16-trimethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate
Figure US09198921-20151201-C00065
EXAMPLE 28 2-((10S,13S,16R,17R)-17-hydroxy-10,13,16-trimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate
Figure US09198921-20151201-C00066
EXAMPLE 29 2-((6S,10R,13S,17R)-17-hydroxy-6,10,13-trimethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate
Figure US09198921-20151201-C00067
EXAMPLE 30 (6S,10R,13S,17R)-17-hydroxy-17-(2-hydroxyacetyl)-6,10,13-trimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one
Figure US09198921-20151201-C00068
EXAMPLE 31 2-((6S,10R,13S,17R)-17-hydroxy-6,10,13-trimethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate
Figure US09198921-20151201-C00069
EXAMPLE 32 (6S,10R,13S,17R)-17-hydroxy-17-(2-hydroxyacetyl)-6,10,13-trimethyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one
Figure US09198921-20151201-C00070
EXAMPLE 33 2-oxo-2-((6S,10R,13S,16R,17S)-6,10,13,16-tetramethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl)ethyl acetate
Figure US09198921-20151201-C00071
EXAMPLE 34 (6S,10R,13S,16R,17S)-17-(2-hydroxyacetyl)-6,10,13,16-tetramethyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-3-one
Figure US09198921-20151201-C00072
EXAMPLE 35 2-oxo-2-((6S,10R,13S,16R,17S)-6,10,13,16-tetramethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)ethyl acetate
Figure US09198921-20151201-C00073
EXAMPLE 36 (6S,10R,13S,16R,17S)-17-(2-hydroxyacetyl)-6,10,13,16-tetramethyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one
Figure US09198921-20151201-C00074
EXAMPLE 37 2-((6S,10R,13S,16R,17R)-17-hydroxy-6,10,13,16-tetramethyl-3-oxo-6,7,8,10,12,13,14,15,16,17-decahydro-3H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate
Figure US09198921-20151201-C00075
EXAMPLE 38 (6S,10R,13S,16R,17R)-17-hydroxy-17-(2-hydroxyacetyl)-6,10,13,16-tetramethyl-6,7,8,10,12,13,14,15,16,17-decahydro-3H -cyclopenta[a]phenanthren-3-one
Figure US09198921-20151201-C00076
EXAMPLE 39 2-((6S,10R,13S,16R,17R)-17-hydroxy-6,10,13,16-tetramethyl-3-oxo-2,3,6,7,8,10,12,13,14,15,16,17-dodecahydro-1H-cyclopenta[a]phenanthren-17-yl)-2-oxoethyl acetate
Figure US09198921-20151201-C00077
EXAMPLE 40 (6S,10R,13S,16R,17R)-17-hydroxy-17-(2-hydroxyacetyl)-6,10,13,16-tetramethyl-6,7,8,10,12,13,14,15,16,17-decahydro-1H-cyclopenta[a]phenanthren-3(2H)-one
Figure US09198921-20151201-C00078
The following compounds can generally be made using the methods described above. It is expected that these compounds when made will have activity similar to those that have been made and tested.
Figure US09198921-20151201-C00079
Figure US09198921-20151201-C00080
Figure US09198921-20151201-C00081
Figure US09198921-20151201-C00082
Figure US09198921-20151201-C00083
Figure US09198921-20151201-C00084
Figure US09198921-20151201-C00085
Figure US09198921-20151201-C00086
Figure US09198921-20151201-C00087
Figure US09198921-20151201-C00088
Figure US09198921-20151201-C00089
Figure US09198921-20151201-C00090
Figure US09198921-20151201-C00091
Figure US09198921-20151201-C00092
Figure US09198921-20151201-C00093
Figure US09198921-20151201-C00094
Figure US09198921-20151201-C00095
The activity of the compounds in Examples 1-7 and 9-10 as NF-κB modulators is illustrated in the following assays. The other compounds listed above, which have not yet been made and/or tested, are predicted to have activity in these assays as well.
Biological Activity Assay
In Vitro NF-kB Inhibitor Screening Assay
C2C12 skeletal muscle cells stably transfected with a luciferase reporter construct regulated under multiple copies of the NF-kB response element (Panomics, Fremont, Calif.) were used to screen NF-kB inhibitors. These cells were maintained at 37° C. with 5% CO2 in a tissue culture incubator with Dulbecco's modified Eagle medium (DMEM) medium containing 10% Fetal bovine serum (FBS) (ATCC, Manassas, Va.), Penicillin 100 U/ml, Streptomycin 100 μg/ml, and 100 μg/ml Hygromysin B (Roche, Indianapolis, Ind.). Screening assays were performed in myoblasts (grown in medium containing 10% FBS) in duplicate 96 well plates at a cell concentration of 5×104 cells per well in 100 ul volume. Cells were pretreated with various concentrations (0.01 ug/ml to 10 ug/ml) of compound for 24 hr duration before stimulating with tumor necrosis factor-α (TNF-α) (10 ng/ml) for another 24 hrs. Prednisolone was included in every plate tested as a positive control. After the completion of incubation cells were washed twice with PBS and lysed with cell lysis buffer to measure luciferase activity (Promega Corp, Madison, Wis.) using Centro LB 960 luminometer (Berthold technologies, GmbH & Co, Bad Wildbad, Germany). Relative luminescence units with TNF-α stimulation in the absence of drugs were considered as 100% percent and data was represented as % inhibition relative to TNF-α induced NF-kB activation.
Some of the compounds disclosed herein were tested in the C2C12 skeletal muscle cell luciferase assay and exhibited ≧100% inhibition at concentrations of 0.01, 0.1, and 1 ug/mL; 80-100% inhibition at concentrations of 0.01, 0.1, 1, and 10 ug/mL; 60-80% inhibition at concentrations of 0.01, 0.1, and 1 ug/mL; 40-60% inhibition at concentrations of 1 and 10 ug/mL; and 20-40% inhibition at concentrations of 10 ug/mL.
Cell viability was assayed in duplicate plates by MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide) (Sigma, Saint Louis, Mo.) as per manufacturer's protocols. Percent cell viability was calculated relative to untreated cells. There was not a significant decrease in cell viability (<80%) for any of compounds tested at any of the doses (0.01, 0.1, 1, and 10 ug/mL) tested.
Inhibition of NF-kB Nuclear Translocation
Inhibition of TNF-α induced NF-kB activation was confirmed by nuclear translocation immunofluorescence assay. C2C12 cells were grown on cover slips and treated with TNF-α and compound at optimal concentrations as described above. Cells were fixed with acetone and stained with a rabbit anti-NF-kB (p60) antibody/anti-rabbit Texas red (Santa Cruz Biotech, Inc, Santa Cruz, and CA) and counterstained with 4′,6-Diamidino-2-phenylindole HCl (DAPI) (Invitrogen, CA) to visualize the nuclei. Some of the compounds disclosed herein were tested in the nuclear translocation immunofluorescence assay and blocked TNF-α induced NF-kB nuclear translocation.
In Vivo mdx Mouse Model of Dystrophy
Separate groups (n=12-14) of mdx mice were treated with prednisolone (5 mg/kg/day; per oral in feed), Example 1 (20 and 40 mg/kg/day; per oral in feed) and Example 2 (20 and 40 mg/kg/day; per oral in feed) for 3 months. All mice underwent 30 min biweekly treadmill exercise during the treatment duration to unmask the mild disease phenotype of mdx mouse model.
Effect on Body Weight (BM)
Prednisolone treated mice showed significantly lower body weight (P<0.05) than untreated mice at 33.8 weeks age. Mice treated with some of the compounds disclosed herein at 20 and 40 mg/kg/day dosages gained significantly more body weight and gastrocnemius muscle mass than untreated mice.
Effect on In Vivo Motor Coordination and Strength
Motor coordination and strength were assessed using Rota-rod (Ugo Basile, VA, Italy) testing. Briefly, mice were trained on the rota-rod for two days before collecting data. Each acclimatization session consisted of four training sessions, 2 per day and each session lasting 120 seconds at a speed of 5 rpm). Each trial consisted of placing the mice on the rod at 10 rpm for 60 seconds (stabilizing period) followed by an acceleration from 10 rpm to 40 rpm within the first 25 seconds until the animal fell from the rod or until 180 seconds are reached. If the animals fell during the stabilizing period, they were placed back on the rod to complete the session. The total testing time was 240 seconds (60 seconds stabilization time and 180 seconds test time). Each trial was performed twice a day (2 hour interval between sessions) for 3 consecutive days. The latency to fall (seconds) was recorded and all six scores were averaged per mouse. The average data was expressed as latency to fall (in seconds) for each group mice at 3 age groups. The ability of untreated mice to stay on the rod did not change significantly with time. Mice treated with some of the compounds disclosed herein at 20 and 40 mg/kg/day dosages showed increased latency to fall at 12, 24, or 36 weeks. In some instances, latency to fall was decreased or unchanged, increased by 0-10%, increased by 10-20%, increased by 20-30%, or increased by 30-40%.
Effect on In Vitro Force Contractions:
The distal tendon of the extensor digitorum longus (EDL) muscle was tied securely to the lever arm of a servomotor/force transducer (model 305B, Aurora Scientific, Richmond Hill, ON, Canada) and the proximal tendon to a tissue clamp. Muscles were stimulated between two platinum electrodes. With supramaximal stimulation of the muscle using single 0.2-ms square stimulation pulses for the EDL, muscle length was adjusted to the length (Lo) that resulted in maximal twitch force. With the muscle held at Lo using stimulation frequencies of 30, 50, 80, 100, 120 and 150 Hz, the maximum isometric tetanic force force (Po) developed during a 300 ms train of stimulation pulses was recorded for the EDL muscle. The muscle length was then measured with calipers and after removal of the muscle from the bath the mass of the muscle was determined. For each muscle, the optimum fiber length (Lf) was calculated by multiplying Lo by a previously determined Lf/Lo ratio of 0.45. Total muscle fiber cross-sectional area was determined by dividing the wet mass by the product of Lf and the density of mammalian skeletal muscle (1.06 mg/mm3). Maximum isometric specific force (sPo) was determined by dividing Po by the total muscle fiber cross-sectional area. There was no statistically significant (P<0.05) difference in specific force between untreated and prednisolone groups. Mice treated with some of the compounds disclosed herein at 20 and 40 mg/kg/day dosages showed statistically significant (P<0.05) increased isometric specific force. In some instances, isometric specific force was decreased or unchanged, increased by 0-5%, increased by 5-10%, increased by 10-15%, or increased by 15-20%.
Histological Evaluations
Hematoxylin and Eosin staining of gastrocnemius muscle of untreated mdx mice show significant degeneration and inflammation. Skeletal muscle from Example 1- and Example 2-treated mice showed significant decrease in inflammation, degeneration, and increase in regenerating muscle fibers in comparison to untreated and prednisolone treated mdx mice. Continuous administration of prednisolone appeared to increase degeneration and decrease in regeneration of dystrophic skeletal muscle.
Glucocorticoid Receptor Binding Assay
To determine the receptor binding affinity of example compounds to the glucocorticoid receptor (GR), a ligand binding assay was performed using cDNA expression clones (Baculovirus) for human and mouse glucocorticoid receptor-alpha. Liver extracts containing different GR constructs were incubated with radiolabeled 3H-Dexamethsone (Amersham Pharmacia Biotech) and test compound in assay buffer (10 mM Tris-HCl, 1.5 mM EDTA, 10% glycerol, 1 mM dithiothreitol, and 20 mM sodium molybdate, pH 7.6). The amount of radioactivity was measured using a scintillation plate reader. Dexamethasone showed competitive binding with 3H-dexamethasone at micromolar concentrations. Some of the compounds disclosed herein were tested in the glucocorticoid receptor binding assay and found to have no significant (>75%) competitive binding to the glucocorticoid receptor at millimolar concentrations.
From the foregoing description, one skilled in the art can easily ascertain the essential characteristics of this invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions.

Claims (48)

What is claimed is:
1. A compound selected from the group consisting of:
Figure US09198921-20151201-C00096
Figure US09198921-20151201-C00097
2. A pharmaceutical composition comprising a compound selected from the group consisting of
Figure US09198921-20151201-C00098
Figure US09198921-20151201-C00099
together with a pharmaceutically acceptable carrier.
3. The pharmaceutical composition as recited in claim 2 wherein said pharmaceutical composition comprises a tablet or a capsule.
4. The pharmaceutical composition as recited in claim 2 wherein said pharmaceutical composition comprises another therapeutic agent.
5. A method of reducing the symptoms of a disease selected from the group consisting of muscular dystrophy, arthritis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type 2 diabetes, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, muscle wasting, catabolic disorders, obesity, fetal growth retardation, hypercholesterolemia, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, asthma, chronic obstructive pulmonary disease, Sjogren's syndrome, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, gut diseases, peritoneal endometriosis, skin diseases, nasal sinusitis, mesothelioma, anhidrotic ecodermal dysplasia-ID, Behcet's disease, incontinentia pigmenti, tuberculosis, asthma, crohn's disease, colitis, ocular allergy, appendicitis, paget's disease, pancreatitis, periodonitis, endometriosis, inflammatory bowel disease, inflammatory lung disease, silica-induced diseases, sleep apnea, AIDS, HIV-1, autoimmune diseases, antiphospholipid syndrome, lupus, lupus nephritis, familial mediterranean fever, hereditary periodic fever syndrome, psychosocial stress diseases, neuropathological diseases, familial amyloidotic polyneuropathy, inflammatory neuropathy, parkinson's disease, multiple sclerosis, alzheimer's disease, amyotropic lateral sclerosis, huntington's disease, cataracts, and hearing loss, comprising the administration, to a patient in need thereof, of a therapeutically effective amount of a compound selected from the group consisting of
Figure US09198921-20151201-C00100
Figure US09198921-20151201-C00101
6. The method as recited in claim 5, wherein said disease is muscular dystrophy.
7. The method as recited in claim 6, wherein said muscular dystrophy is selected from the group consisting of Duchenne muscular dystrophy, Becker muscular dystrophy, limb girdle muscular dystrophy, congenital muscular dystrophy, facioscapulohumeral muscular dystrophy, myotonic muscular dystrophy, oculopharyngeal muscular dystrophy, distal muscular dystrophy, and Emery-Dreifuss muscular dystrophy.
8. The method as recited in claim 7, wherein said muscular dystrophy is Duchenne muscular dystrophy.
9. The method as recited in claim 5, wherein said disease is selected from the group consisting of asthma and chronic obstructive pulmonary disease.
10. The method as recited in claim 9, wherein said disease is asthma.
11. The method as recited in claim 9, wherein said disease is chronic obstructive pulmonary disease.
12. The method as recited in claim 5, wherein said disease is arthritis.
13. The method as recited in claim 5, wherein said disease is selected from the group consisting of traumatic brain injury and spinal cord injury.
14. The method as recited in claim 13, wherein said disease is traumatic brain injury.
15. The method as recited in claim 13, wherein said disease is spinal cord injury.
16. The method as recited in claim 5, wherein said disease is amyotropic lateral sclerosis.
17. The method as recited in claim 5, wherein said disease is Sjogren's syndrome.
18. The method as recited in claim 5, wherein said compound has the structural formula:
Figure US09198921-20151201-C00102
19. The method as recited in claim 5, wherein said compound has the structural formula:
Figure US09198921-20151201-C00103
20. The method as recited in claim 5, wherein said compound has the structural formula:
Figure US09198921-20151201-C00104
21. The method as recited in claim 5, wherein said compound has the structural formula:
Figure US09198921-20151201-C00105
22. The method as recited in claim 5, wherein said compound has the structural formula:
Figure US09198921-20151201-C00106
23. The method as recited in claim 5, wherein said compound has the structural formula:
Figure US09198921-20151201-C00107
24. The method as recited in claim 5, wherein said compound has the structural formula:
Figure US09198921-20151201-C00108
25. The method as recited in claim 5, wherein said compound has the structural formula:
Figure US09198921-20151201-C00109
26. The method as recited in claim 5, comprising the administration of another therapeutic agent.
27. A method of reducing the symptoms of a disease selected from the group consisting of muscular dystrophy, arthritis, traumatic brain injury, spinal cord injury, sepsis, rheumatic disease, cancer atherosclerosis, type 1 diabetes, type 2 diabetes, leptospiriosis renal disease, glaucoma, retinal disease, ageing, headache, pain, complex regional pain syndrome, cardiac hypertrophy, muscle wasting, catabolic disorders, obesity, fetal growth retardation, hypercholesterolemia, heart disease, chronic heart failure, ischemia/reperfusion, stroke, cerebral aneurysm, angina pectoris, pulmonary disease, cystic fibrosis, acid-induced lung injury, pulmonary hypertension, asthma, chronic obstructive pulmonary disease, Sjogren's syndrome, hyaline membrane disease, kidney disease, glomerular disease, alcoholic liver disease, gut diseases, peritoneal endometriosis, skin diseases, nasal sinusitis, mesothelioma, anhidrotic ecodermal dysplasia-ID, Behcet's disease, incontinentia pigmenti, tuberculosis, asthma, crohn's disease, colitis, ocular allergy, appendicitis, paget's disease, pancreatitis, periodonitis, endometriosis, inflammatory bowel disease, inflammatory lung disease, silica-induced diseases, sleep apnea, AIDS, HIV-1, autoimmune diseases, antiphospholipid syndrome, lupus, lupus nephritis, familial mediterranean fever, hereditary periodic fever syndrome, psychosocial stress diseases, neuropathological diseases, familial amyloidotic polyneuropathy, inflammatory neuropathy, parkinson's disease, multiple sclerosis, alzheimer's disease, amyotropic lateral sclerosis, huntington's disease, cataracts, and hearing loss, comprising the administration, to a patient in need thereof, of a therapeutically effective amount of a compound selected from the group consisting of
Figure US09198921-20151201-C00110
Figure US09198921-20151201-C00111
28. The method as recited in claim 27, wherein said disease is muscular dystrophy.
29. The method as recited in claim 28, wherein said muscular dystrophy is selected from the group consisting of Duchenne muscular dystrophy, Becker muscular dystrophy, limb girdle muscular dystrophy, congenital muscular dystrophy, facioscapulohumeral muscular dystrophy, myotonic muscular dystrophy, oculopharyngeal muscular dystrophy, distal muscular dystrophy, and Emery-Dreifuss muscular dystrophy.
30. The method as recited in claim 29, wherein said muscular dystrophy is Duchenne muscular dystrophy.
31. The method as recited in claim 27, wherein said disease is selected from the group consisting of asthma and chronic obstructive pulmonary disease.
32. The method as recited in claim 31, wherein said disease is asthma.
33. The method as recited in claim 31, wherein said disease is chronic obstructive pulmonary disease.
34. The method as recited in claim 27, wherein said disease is arthritis.
35. The method as recited in claim 27, wherein said disease is selected from the group consisting of traumatic brain injury and spinal cord injury.
36. The method as recited in claim 35, wherein said disease is traumatic brain injury.
37. The method as recited in claim 35, wherein said disease is spinal cord injury.
38. The method as recited in claim 27, wherein said disease is amyotropic lateral sclerosis.
39. The method as recited in claim 27, wherein said disease is Sjogren's syndrome.
40. The method as recited in claim 27, wherein said compound has the structural formula:
Figure US09198921-20151201-C00112
41. The method as recited in claim 27, wherein said compound has the structural formula:
Figure US09198921-20151201-C00113
42. The method as recited in claim 27, wherein said compound has the structural formula:
Figure US09198921-20151201-C00114
43. The method as recited in claim 27, wherein said compound has the structural formula:
Figure US09198921-20151201-C00115
44. The method as recited in claim 27, wherein said compound has the structural formula:
Figure US09198921-20151201-C00116
45. The method as recited in claim 27, wherein said compound has the structural formula:
Figure US09198921-20151201-C00117
46. The method as recited in claim 27, wherein said compound has the structural formula:
Figure US09198921-20151201-C00118
47. The method as recited in claim 27, wherein said compound has the structural formula:
Figure US09198921-20151201-C00119
48. The method as recited in claim 27, comprising the administration of another therapeutic agent.
US13/639,650 2010-04-05 2011-04-05 Non-hormonal steroid modulators of NF-κB for treatment of disease Active US9198921B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/639,650 US9198921B2 (en) 2010-04-05 2011-04-05 Non-hormonal steroid modulators of NF-κB for treatment of disease

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US32090510P 2010-04-05 2010-04-05
US61320905 2010-04-05
US13/639,650 US9198921B2 (en) 2010-04-05 2011-04-05 Non-hormonal steroid modulators of NF-κB for treatment of disease
PCT/US2011/031238 WO2011127048A2 (en) 2010-04-05 2011-04-05 NON-HORMONAL STEROID MODULATORS OF NF-ĸB FOR TREATMENT OF DISEASE

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/031238 A-371-Of-International WO2011127048A2 (en) 2010-04-05 2011-04-05 NON-HORMONAL STEROID MODULATORS OF NF-ĸB FOR TREATMENT OF DISEASE

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/939,717 Continuation US10000525B2 (en) 2010-04-05 2015-11-12 Non-hormonal steroid modulators of NF-κB for treatment of disease

Publications (2)

Publication Number Publication Date
US20130196962A1 US20130196962A1 (en) 2013-08-01
US9198921B2 true US9198921B2 (en) 2015-12-01

Family

ID=44763515

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/639,650 Active US9198921B2 (en) 2010-04-05 2011-04-05 Non-hormonal steroid modulators of NF-κB for treatment of disease
US14/939,717 Active US10000525B2 (en) 2010-04-05 2015-11-12 Non-hormonal steroid modulators of NF-κB for treatment of disease

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/939,717 Active US10000525B2 (en) 2010-04-05 2015-11-12 Non-hormonal steroid modulators of NF-κB for treatment of disease

Country Status (3)

Country Link
US (2) US9198921B2 (en)
EP (1) EP2556083A4 (en)
WO (1) WO2011127048A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160375037A1 (en) * 2015-06-29 2016-12-29 Reveragen Biopharma, Inc. Non-hormonal steroid modulators of nf-kappa beta for treatment of disease
WO2020191087A1 (en) 2019-03-18 2020-09-24 Ptc Therapeutics, Inc. Therapeutic combinations for use in treating a muscular dystrophy
US10857161B2 (en) 2008-05-28 2020-12-08 Reveragen Biopharma, Inc. Non-hormonal steroid modulators of NF-kB for treatment of disease
US11382922B2 (en) 2019-03-07 2022-07-12 Reveragen Biopharma, Inc. Aqueous oral pharmaceutical suspension compositions
EP4230196A1 (en) 2022-02-21 2023-08-23 Som Innovation Biotech, S.A. Compounds for use in the treatment of dystrophinopathies

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9198921B2 (en) 2010-04-05 2015-12-01 Reveragen Biopharma, Inc. Non-hormonal steroid modulators of NF-κB for treatment of disease
CA2855756A1 (en) * 2011-11-29 2013-06-06 Reveragen Biopharma, Inc. Non-hormonal steroid modulators of nf-kb for treatment of disease
WO2014169126A1 (en) 2013-04-10 2014-10-16 Reveragen Biopharma, Inc. Methods and agents to increase therapeutic dystrophin expression in muscle
CN107286216A (en) * 2016-03-31 2017-10-24 天津金耀集团有限公司 Preparation method with multiple alkene steroidal compounds

Citations (113)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2776927A (en) 1955-12-27 1957-01-08 Pfizer & Co C Preparation of delta 1, 4-3-keto steroids from delta4 3-keto steroids by protaminobacter
US2838536A (en) 1957-11-29 1958-06-10 Upjohn Co 6-fluoro 9 alpha-halo-delta4-pregnenes and process for preparing same
GB814000A (en) 1954-05-21 1959-05-27 Pfizer & Co C Steroid compounds and the preparation thereof
US2894963A (en) 1958-06-20 1959-07-14 Scherico Ltd 9,11-dihalogeno-4-pregnenes and 1,4-pregnadienes
GB843214A (en) 1955-01-11 1960-08-04 Schering Corp Improvements in or relating to steroid compounds
GB843215A (en) 1954-10-05 1960-08-04 Schering Corp Improvements in or relating to steroid compounds
US2957893A (en) 1955-01-20 1960-10-25 Schering Corp delta1, 4, 9(11)-pregnatrienes
US2980713A (en) 1958-09-05 1961-04-18 Merck & Co Inc 16-methyl-3alpha, 17alpha-dihydroxy steroids of the pregnane series
US3004965A (en) 1959-08-07 1961-10-17 Smith Kline French Lab Process and intermediates for preparing steroidal 17->18 lactones
US3009933A (en) 1959-06-01 1961-11-21 Schering Corp Process for the manufacture of 9alpha, 11beta-dihalogenated steroids
GB901093A (en) 1957-07-22 1962-07-11 Scherico Ltd Steroids
US3047468A (en) 1961-06-06 1962-07-31 American Cyanamid Co Method of preparing delta1, 4-3-keto steroids of the pregnane series
US3053866A (en) 1961-03-02 1962-09-11 Merck & Co Inc Improved steroid dehydrogenation process
GB912378A (en) 1958-06-19 1962-12-05 Merck & Co Inc Steroid compounds
US3087938A (en) 1961-08-02 1963-04-30 Schering Corp 17alpha-chloro and 17alpha-bromo-progesterones
GB928301A (en) 1958-06-20 1963-06-12 Scherico Ltd 9, 11-dihalogeno steroids
US3098086A (en) 1959-01-22 1963-07-16 Ciba Geigy Corp Process for the manufacture of 11beta-hydroxy-pregnane compounds
US3127425A (en) 1961-05-29 1964-03-31 Schering Corp 6, 17alpha-disubstituted 9alpha, 11beta-dihalogeno derivatives of a-ring unsaturated pregnane 3, 20-diones
GB959378A (en) 1959-10-12 1964-06-03 Scherico Ltd 21-substituted 17ª‡, 21-dioxy steroids and processes for their preparation
FR1433301A (en) 1959-06-12 1966-04-01 Merck & Co Inc Process for the preparation of 16alpha-methyl, 1, 4-pregnadiene compounds
US3284477A (en) 1963-07-10 1966-11-08 Schering Corp Diuretic 1, 4, 9(11)-pregnatrienes
US3463852A (en) 1966-01-14 1969-08-26 Schering Corp Treating allergies with steroids of the pregnane series
US3681405A (en) 1969-07-24 1972-08-01 Schering Ag Novel 21,21-dichlorosteroids
US3842105A (en) 1972-02-11 1974-10-15 Schering Ag Process for the preparation of 16,17-unsaturated steroids
US3891631A (en) 1972-08-11 1975-06-24 Glaxo Lab Ltd Process for preparing 17{60 -monoesters of 17{60 , 21-dihydroxy-20-oxo steroids
CA975755A (en) 1971-05-07 1975-10-07 Jozsef Toth Nitric acid esters of 21-alcohols belonging to the pregnane series as well as a process for the preparation thereof
US3923985A (en) 1973-07-27 1975-12-02 Roussel Uclaf Process for the preparation of 16{60 -methyl-{66 {hu 1,4,9(11){b -pregnatrien-3,20-diones and use of the same in treatment of allergies
CA983919A (en) 1971-07-05 1976-02-17 Gordon H. Phillipps Steroid compounds
ES445981A1 (en) 1976-03-11 1977-06-01 Aldo Union Calzada Y Cia S R C Procedure for the preparation of an intermediate compound in the synthesis of 9 alpha-halogen-16 beta-methyl-prednisolone. (Machine-translation by Google Translate, not legally binding)
US4041055A (en) 1975-11-17 1977-08-09 The Upjohn Company Process for the preparation of 17α-hydroxyprogesterones and corticoids from androstenes
US4076708A (en) 1976-12-22 1978-02-28 Schering Corporation Process for the preparation of 7α-halogeno-3-oxo-4-dehydro steroids and novel 7α-halogeno derivatives produced thereby
CA1086715A (en) 1976-06-04 1980-09-30 Henry L. Levinsky Steroids
US4260464A (en) 1977-10-26 1981-04-07 Schering Aktiengesellschaft Process for the preparation of 17α-(3-iodobenzoyloxy)-9α-chloro-4-pregnene-3,20-diones and their D-homo homologs
US4318853A (en) 1980-01-31 1982-03-09 The Upjohn Company 9β,11β-Epoxy-5β-corticoids
US4336200A (en) 1980-01-31 1982-06-22 The Upjohn Company 17α-Acyloxy-5β-corticoids
GB2089808A (en) 1980-12-22 1982-06-30 Upjohn Co 5 alpha - and 5 beta -androstane-17 beta - carboxylates and -thiocarboxylates
EP0057401A1 (en) 1981-02-02 1982-08-11 Schering Corporation Aromatic heterocyclic esters of steroids, their preparation and pharmaceutical compositions containing them
US4404141A (en) 1981-03-09 1983-09-13 Schering Aktiengesellschaft Δ9(11) - And Δ16 -21-chloro-20-keto steroids of the pregnane and D-homopregnane series, their preparation and use as intermediates for the synthesis of highly effective corticoids
DE3227312A1 (en) 1982-07-19 1984-01-19 Schering AG, 1000 Berlin und 4709 Bergkamen NEW 6.16 DIMETHYL CORTICOIDS, THEIR PRODUCTION AND USE
US4427591A (en) 1980-01-31 1984-01-24 The Upjohn Company Reduced A ring-Δ9(11) -corticoids
US4444689A (en) 1980-01-31 1984-04-24 The Upjohn Company 17α-Acyloxy-5β-corticoids and 17α-acyloxy-5α-corticoids
GB2131811A (en) 1982-07-30 1984-06-27 Upjohn Co Microbial 1,2-dehydrogenation of steroids
EP0114589A1 (en) 1982-12-20 1984-08-01 The President And Fellows Of Harvard College Inhibition of angiogenesis
EP0126877A1 (en) 1983-04-27 1984-12-05 Schering Aktiengesellschaft Process for the preparation of 3-keto-delta-1,4-steroids
EP0156643A2 (en) 1984-03-28 1985-10-02 The Upjohn Company Ester prodrugs of steroids
EP0164298A2 (en) 1984-03-13 1985-12-11 Schering Aktiengesellschaft 6-Alpha-methyl-D-homo-corticoids
US4613463A (en) 1984-11-01 1986-09-23 The Upjohn Company Process and intermediates for the preparation of 17 alphahydroxyprogesterones and corticoids from an enol steroid
US4645763A (en) 1982-11-22 1987-02-24 Schering Aktiengesellschaft 6α-methyl corticoids, their production and use
WO1987001706A2 (en) 1985-09-12 1987-03-26 The Upjohn Company C20 through c26 amino steroids
WO1987007895A1 (en) 1986-06-23 1987-12-30 The Upjohn Company Androstane-type and cortical aminoesters
EP0263213A1 (en) 1986-10-09 1988-04-13 The Upjohn Company C20 Through C26 amino steroids
US4771042A (en) 1985-11-25 1988-09-13 The Upjohn Company Inhibition of angiogenesis involving the coadministration of steroids with heparin or heparin fragments
WO1988007092A1 (en) 1987-03-12 1988-09-22 The Upjohn Company 1,2-dehydrogenation of steroidal 21-esters with a. simplex
WO1988007527A1 (en) 1987-04-03 1988-10-06 The Upjohn Company Amino-9,10-secosteroids
US4777165A (en) 1984-01-16 1988-10-11 Schering Aktiengesellschaft 6α, 16β-dimethyl corticoids and their anti-inflammatory use
US4876250A (en) 1988-10-31 1989-10-24 Alcon Laboratories, Inc. Methods for controlling ocular hypertension with angiostatic steroids
US4891426A (en) 1984-06-11 1990-01-02 The Upjohn Company 16α-methyl-17α 20-epoxysteroid and process
US4910192A (en) 1987-12-04 1990-03-20 Sri International Topically active steroidal anti-inflammatory agents
US4920216A (en) 1987-05-28 1990-04-24 The Trustees Of Columbia In The City Of New York Selective chlorination of steroids and other substrates directed by covalently linked pyridine derivatives acting as templates
US4929395A (en) 1984-06-11 1990-05-29 The Upjohn Company 16α-methylation process
US4948533A (en) 1984-03-28 1990-08-14 The Upjohn Company 11a-hydroxy steroid diester
US4975537A (en) 1985-10-23 1990-12-04 The Upjohn Company Δ9(11) -angiostatic steroids
US4977255A (en) 1986-11-05 1990-12-11 The Upjohn Company Steroidal 17α-silyl ethers and process to corticoids and progesterones
WO1990015816A1 (en) 1989-06-16 1990-12-27 The Upjohn Company Suramin type compounds and angiostatic steroids to inhibit angiogenesis
US4994443A (en) 1982-12-20 1991-02-19 The Children's Medical Center Corporation Inhibition of angiogenesis
US5001116A (en) 1982-12-20 1991-03-19 The Children's Medical Center Corporation Inhibition of angiogenesis
WO1991019731A1 (en) 1990-06-11 1991-12-26 The Upjohn Company Steroids which inhibit angiogenesis
WO1990012577A3 (en) 1989-04-18 1992-03-19 Upjohn Co Topical anti-angiogenic as hair growth inhibitors
US5248773A (en) 1991-06-25 1993-09-28 Roussel-Uclaf 16-methyl-Δ1,4-pregnadiene-3,20-diones
WO1993025570A1 (en) 1992-06-09 1993-12-23 Richter Gedeon Vegyészeti Gyár Rt. Novel steroids with pregnane skeleton, pharmaceutical compositions containing them and process for preparing same
US5310896A (en) 1991-04-10 1994-05-10 Roussel Uclaf Process for preparing Δ1,4 -pregnadiene-3,20-diones
US5412091A (en) 1991-06-25 1995-05-02 Roussel Uclaf 16-methyl-Δ1,4-pregnadiene-3,20-dione
WO1995018621A1 (en) 1994-01-04 1995-07-13 Duke University Methods of inhibiting angiogenesis and tumor growth, and treating ophthalmologic conditions with angiostatic and therapeutic steroids
WO1996006618A1 (en) 1994-09-01 1996-03-07 Pharmacia & Upjohn Company Cosolvent parenteral formulation of tirilazad
US5502222A (en) 1994-06-01 1996-03-26 Schering Corporation Process for preparing delta 9,11 and 21-chloro corticosteroids
US5502183A (en) 1992-01-28 1996-03-26 Schering Corporation Steroid intermediates and processes for their preparation
WO1996010402A1 (en) 1994-10-04 1996-04-11 Knox Van Dyke Multi-faceted method to repress reproduction of latent viruses in humans and animals
US5508452A (en) 1993-01-20 1996-04-16 Roussel Uclaf Preparation of 16α-methyl steroids
WO1997022616A1 (en) 1995-12-20 1997-06-26 Schering Corporation PROCESS FOR PREPARATION OF 9,11β-EPOXIDE STEROIDS
WO1997039018A1 (en) 1996-04-18 1997-10-23 Hoechst Marion Roussel New pregnane derivatives with no alpha-17 substitutent, their medicinal use, manufacturing method and its intermediaries and related compounds
US5731447A (en) 1992-06-11 1998-03-24 Roussel Uclaf Process for preparing 16α-methyl-steroids
EP0875516A2 (en) 1997-04-30 1998-11-04 Farmabios S.r.l. Process for the preparation of 16,17 acetals of pregnane derivatives with control of the epimeric distribution at the C-22 position
WO1999003503A1 (en) 1997-07-16 1999-01-28 Alcon Laboratories, Inc. The use of angiostatic steroids in photodynamic therapy
WO1999017778A1 (en) 1997-10-07 1999-04-15 Cephalon Inc. α-KETOAMIDE MULTICATALYTIC PROTEASE INHIBITORS
WO1999032102A1 (en) 1997-12-19 1999-07-01 Alcon Laboratories, Inc. Treatment of glc1a glaucoma with glucocorticoid antagonists
US5972922A (en) 1990-06-11 1999-10-26 Alcon Laboratories, Inc. Steroids which inhibit angiogenesis
US5990099A (en) 1988-10-31 1999-11-23 Alcon Laboratories, Inc. Angiostatic agents and methods and compositions for controlling ocular hypertension
US6011023A (en) 1997-08-27 2000-01-04 Alcon Laboratories, Inc. Angiostatic steroids
WO2000038653A1 (en) 1998-12-23 2000-07-06 Idea Ag. Improved formulation for topical non-invasive application in vivo
US6090794A (en) 1990-04-19 2000-07-18 The General Hospital Corporation Inhibition of neurofibrosarcoma growth and angiogenesis
RU2156256C1 (en) 1999-12-20 2000-09-20 Андрюшина Валентина Александровна Method of preparing corticosteroids
WO2002043785A2 (en) 2000-11-29 2002-06-06 Oculex Pharmaceuticals, Inc. Intraocular implants for preventing transplant rejection in the eye
EP1236469A2 (en) 1991-11-22 2002-09-04 Alcon Laboratories, Inc. Angiostatic steroids
US20030032630A1 (en) 1998-05-27 2003-02-13 Manuel Merlos Roca Use of a 2-hydroxy-4-trifluoromethylbenzoic acid derivatives as inhibitors of the activation of the nuclear transcription factor NF-kappaB
WO2003014141A1 (en) 2001-08-10 2003-02-20 Cosmo S.P.A. 17alfa, 21-dihydroxypregnene esters as antiandrogenic agents
EP1336602A1 (en) 2002-02-13 2003-08-20 Giovanni Scaramuzzino Nitrate prodrugs able to release nitric oxide in a controlled and selective way and their use for prevention and treatment of inflammatory, ischemic and proliferative diseases
WO2004012699A2 (en) 2002-08-05 2004-02-12 Torrent Pharmaceuticals Limited Modified release composition comprising coated micro matrix particles containing the high soluble active ingredient and a release controlling agent
WO2004012700A2 (en) 2002-08-05 2004-02-12 Torrent Pharmaceuticals Limited Dosage form comprising high dose high soluble active ingredient as modified release and low dose active ingredient as immediate release
US20040181055A1 (en) 2001-07-26 2004-09-16 Ragactives, S.L. Stereoselective process for the production of 6alpha-fluorpregnanes and intermediates
US20050090553A1 (en) 1992-06-30 2005-04-28 Shapiro Howard K. Compositions and method for treatment of chronic inflammatory diseases
CN1896090A (en) 2005-07-14 2007-01-17 天津药业研究院有限公司 Preparation of delta 9(11) steroid compound
CN1907999A (en) 2005-08-05 2007-02-07 天津天药药业股份有限公司 Preparation method for 9,11-de-esterification of metllylpredllisolone series products
US20070212751A1 (en) 2006-01-18 2007-09-13 Solvay Pharmaceuticals Gmbh Microbial method for the 11beta hydroxylation of 9beta, 10alpha-steriods
US20080234380A1 (en) 1992-06-30 2008-09-25 Shapiro Howard K Compositions and method for treatment of chronic inflammatory diseases
CN101347438A (en) 2007-07-20 2009-01-21 天津药业研究院有限公司 Medicament for slowing down angiogenesis of eye section
CN101353368A (en) 2007-07-27 2009-01-28 天津药业研究院有限公司 Pregna medicament for treating tumor
CN101397320A (en) 2007-09-29 2009-04-01 天津天药药业股份有限公司 Method for preparing dexamethasone and series products thereof
CN101397319A (en) 2007-09-29 2009-04-01 天津天药药业股份有限公司 Method for preparing betamethasone and series products thereof
CN101434631A (en) 2007-11-16 2009-05-20 天津金耀集团有限公司 Estra nitrate ester medicament for inhibiting angiogenesis
WO2009155056A2 (en) 2008-05-28 2009-12-23 Validus Genetics Non-hormonal steroid modulators of nf-kb for treatment of disease
CN101624414A (en) 2008-07-07 2010-01-13 天津金耀集团有限公司 Nitrate medicament for inhibiting angiogenesis
CN101759742A (en) 2008-11-06 2010-06-30 天津金耀集团有限公司 Preparation method of 16 Alpha-methyl steroidal compound
WO2011127048A2 (en) 2010-04-05 2011-10-13 Validus Biopharma NON-HORMONAL STEROID MODULATORS OF NF-ĸB FOR TREATMENT OF DISEASE

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2928852A (en) * 1958-03-19 1960-03-15 Upjohn Co 6-methyl-17alpha, 21-dihydroxy-4, 9(11)-pregnadiene-3, 20-dione and the 21-esters thereof
GB869511A (en) * 1958-05-26 1961-05-31 Upjohn Co Improvements in or relating to steroids and the manufacture thereof
US3094543A (en) * 1961-07-03 1963-06-18 Merck & Co Inc Preparation of 9 (11)-anhydro steroids from 11-hydroxy steroids
DE2059310C3 (en) * 1970-11-28 1980-10-30 Schering Ag medicaments containing them and processes for their production
GB1509713A (en) 1974-09-21 1978-05-04 Communications Patents Ltd Switches
US4762542A (en) 1987-03-20 1988-08-09 The Boc Group, Inc. Process for the recovery of argon

Patent Citations (139)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB814000A (en) 1954-05-21 1959-05-27 Pfizer & Co C Steroid compounds and the preparation thereof
GB843215A (en) 1954-10-05 1960-08-04 Schering Corp Improvements in or relating to steroid compounds
GB843214A (en) 1955-01-11 1960-08-04 Schering Corp Improvements in or relating to steroid compounds
US2957893A (en) 1955-01-20 1960-10-25 Schering Corp delta1, 4, 9(11)-pregnatrienes
US2776927A (en) 1955-12-27 1957-01-08 Pfizer & Co C Preparation of delta 1, 4-3-keto steroids from delta4 3-keto steroids by protaminobacter
GB901093A (en) 1957-07-22 1962-07-11 Scherico Ltd Steroids
US2838536A (en) 1957-11-29 1958-06-10 Upjohn Co 6-fluoro 9 alpha-halo-delta4-pregnenes and process for preparing same
GB912378A (en) 1958-06-19 1962-12-05 Merck & Co Inc Steroid compounds
US2894963A (en) 1958-06-20 1959-07-14 Scherico Ltd 9,11-dihalogeno-4-pregnenes and 1,4-pregnadienes
GB928301A (en) 1958-06-20 1963-06-12 Scherico Ltd 9, 11-dihalogeno steroids
GB928302A (en) 1958-06-20 1963-06-12 Scherico Ltd í¸-pregnene compounds
US2980713A (en) 1958-09-05 1961-04-18 Merck & Co Inc 16-methyl-3alpha, 17alpha-dihydroxy steroids of the pregnane series
US3098086A (en) 1959-01-22 1963-07-16 Ciba Geigy Corp Process for the manufacture of 11beta-hydroxy-pregnane compounds
US3009933A (en) 1959-06-01 1961-11-21 Schering Corp Process for the manufacture of 9alpha, 11beta-dihalogenated steroids
FR1433301A (en) 1959-06-12 1966-04-01 Merck & Co Inc Process for the preparation of 16alpha-methyl, 1, 4-pregnadiene compounds
US3004965A (en) 1959-08-07 1961-10-17 Smith Kline French Lab Process and intermediates for preparing steroidal 17->18 lactones
GB959378A (en) 1959-10-12 1964-06-03 Scherico Ltd 21-substituted 17ª‡, 21-dioxy steroids and processes for their preparation
US3053866A (en) 1961-03-02 1962-09-11 Merck & Co Inc Improved steroid dehydrogenation process
US3127425A (en) 1961-05-29 1964-03-31 Schering Corp 6, 17alpha-disubstituted 9alpha, 11beta-dihalogeno derivatives of a-ring unsaturated pregnane 3, 20-diones
US3047468A (en) 1961-06-06 1962-07-31 American Cyanamid Co Method of preparing delta1, 4-3-keto steroids of the pregnane series
US3087938A (en) 1961-08-02 1963-04-30 Schering Corp 17alpha-chloro and 17alpha-bromo-progesterones
US3284477A (en) 1963-07-10 1966-11-08 Schering Corp Diuretic 1, 4, 9(11)-pregnatrienes
US3463852A (en) 1966-01-14 1969-08-26 Schering Corp Treating allergies with steroids of the pregnane series
US3681405A (en) 1969-07-24 1972-08-01 Schering Ag Novel 21,21-dichlorosteroids
CA975755A (en) 1971-05-07 1975-10-07 Jozsef Toth Nitric acid esters of 21-alcohols belonging to the pregnane series as well as a process for the preparation thereof
CA983919A (en) 1971-07-05 1976-02-17 Gordon H. Phillipps Steroid compounds
US3842105A (en) 1972-02-11 1974-10-15 Schering Ag Process for the preparation of 16,17-unsaturated steroids
US3891631A (en) 1972-08-11 1975-06-24 Glaxo Lab Ltd Process for preparing 17{60 -monoesters of 17{60 , 21-dihydroxy-20-oxo steroids
US3923985A (en) 1973-07-27 1975-12-02 Roussel Uclaf Process for the preparation of 16{60 -methyl-{66 {hu 1,4,9(11){b -pregnatrien-3,20-diones and use of the same in treatment of allergies
GB1480763A (en) 1973-07-27 1977-07-27 Roussel Uclaf Pharmaceutical compositions containing steroids
US4041055A (en) 1975-11-17 1977-08-09 The Upjohn Company Process for the preparation of 17α-hydroxyprogesterones and corticoids from androstenes
ES445981A1 (en) 1976-03-11 1977-06-01 Aldo Union Calzada Y Cia S R C Procedure for the preparation of an intermediate compound in the synthesis of 9 alpha-halogen-16 beta-methyl-prednisolone. (Machine-translation by Google Translate, not legally binding)
CA1086715A (en) 1976-06-04 1980-09-30 Henry L. Levinsky Steroids
US4076708A (en) 1976-12-22 1978-02-28 Schering Corporation Process for the preparation of 7α-halogeno-3-oxo-4-dehydro steroids and novel 7α-halogeno derivatives produced thereby
US4260464A (en) 1977-10-26 1981-04-07 Schering Aktiengesellschaft Process for the preparation of 17α-(3-iodobenzoyloxy)-9α-chloro-4-pregnene-3,20-diones and their D-homo homologs
US4318853A (en) 1980-01-31 1982-03-09 The Upjohn Company 9β,11β-Epoxy-5β-corticoids
US4336200A (en) 1980-01-31 1982-06-22 The Upjohn Company 17α-Acyloxy-5β-corticoids
US4427591A (en) 1980-01-31 1984-01-24 The Upjohn Company Reduced A ring-Δ9(11) -corticoids
US4444689A (en) 1980-01-31 1984-04-24 The Upjohn Company 17α-Acyloxy-5β-corticoids and 17α-acyloxy-5α-corticoids
GB2089808A (en) 1980-12-22 1982-06-30 Upjohn Co 5 alpha - and 5 beta -androstane-17 beta - carboxylates and -thiocarboxylates
DE3149475A1 (en) 1980-12-22 1982-07-15 The Upjohn Co., 49001 Kalamazoo, Mich. 5 (ALPHA) ANDROSTAN-17 (BETA) CARBOXYLATE AND 5 (BETA) ANDROSTAN-17 (BETA) CARBOXYLATE
US4472393A (en) 1981-02-02 1984-09-18 Schering Corporation 3,20-Dioxo-1,4-pregnadiene-17α-ol 17-aromatic heterocycle carboxylates
EP0057401A1 (en) 1981-02-02 1982-08-11 Schering Corporation Aromatic heterocyclic esters of steroids, their preparation and pharmaceutical compositions containing them
US4404141A (en) 1981-03-09 1983-09-13 Schering Aktiengesellschaft Δ9(11) - And Δ16 -21-chloro-20-keto steroids of the pregnane and D-homopregnane series, their preparation and use as intermediates for the synthesis of highly effective corticoids
US4555507A (en) 1982-07-19 1985-11-26 Schering Aktiengesellschaft Novel 6, 16-dimethylcorticoids, their preparation and use
US4701451A (en) 1982-07-19 1987-10-20 Schering Aktiengesellschaft Novel 6,16-dimethylcorticoids their preparation and use
DE3227312A1 (en) 1982-07-19 1984-01-19 Schering AG, 1000 Berlin und 4709 Bergkamen NEW 6.16 DIMETHYL CORTICOIDS, THEIR PRODUCTION AND USE
GB2131811A (en) 1982-07-30 1984-06-27 Upjohn Co Microbial 1,2-dehydrogenation of steroids
US4645763A (en) 1982-11-22 1987-02-24 Schering Aktiengesellschaft 6α-methyl corticoids, their production and use
EP0114589A1 (en) 1982-12-20 1984-08-01 The President And Fellows Of Harvard College Inhibition of angiogenesis
US4994443A (en) 1982-12-20 1991-02-19 The Children's Medical Center Corporation Inhibition of angiogenesis
US5001116A (en) 1982-12-20 1991-03-19 The Children's Medical Center Corporation Inhibition of angiogenesis
EP0126877A1 (en) 1983-04-27 1984-12-05 Schering Aktiengesellschaft Process for the preparation of 3-keto-delta-1,4-steroids
US4777165A (en) 1984-01-16 1988-10-11 Schering Aktiengesellschaft 6α, 16β-dimethyl corticoids and their anti-inflammatory use
EP0164298A2 (en) 1984-03-13 1985-12-11 Schering Aktiengesellschaft 6-Alpha-methyl-D-homo-corticoids
US4948533A (en) 1984-03-28 1990-08-14 The Upjohn Company 11a-hydroxy steroid diester
EP0156643A2 (en) 1984-03-28 1985-10-02 The Upjohn Company Ester prodrugs of steroids
US4990612A (en) 1984-06-11 1991-02-05 The Upjohn Company 16α-methylation process
US4891426A (en) 1984-06-11 1990-01-02 The Upjohn Company 16α-methyl-17α 20-epoxysteroid and process
US4929395A (en) 1984-06-11 1990-05-29 The Upjohn Company 16α-methylation process
US4613463A (en) 1984-11-01 1986-09-23 The Upjohn Company Process and intermediates for the preparation of 17 alphahydroxyprogesterones and corticoids from an enol steroid
WO1987001706A2 (en) 1985-09-12 1987-03-26 The Upjohn Company C20 through c26 amino steroids
US4975537A (en) 1985-10-23 1990-12-04 The Upjohn Company Δ9(11) -angiostatic steroids
US4771042A (en) 1985-11-25 1988-09-13 The Upjohn Company Inhibition of angiogenesis involving the coadministration of steroids with heparin or heparin fragments
WO1987007895A1 (en) 1986-06-23 1987-12-30 The Upjohn Company Androstane-type and cortical aminoesters
EP0263213A1 (en) 1986-10-09 1988-04-13 The Upjohn Company C20 Through C26 amino steroids
US4977255A (en) 1986-11-05 1990-12-11 The Upjohn Company Steroidal 17α-silyl ethers and process to corticoids and progesterones
WO1988007092A1 (en) 1987-03-12 1988-09-22 The Upjohn Company 1,2-dehydrogenation of steroidal 21-esters with a. simplex
US5225335A (en) 1987-03-12 1993-07-06 The Upjohn Company 1,2-dehydrogenation of steroidal 21-esters with Arthrobacter simplex or Bacterium cyclooxydans
WO1988007527A1 (en) 1987-04-03 1988-10-06 The Upjohn Company Amino-9,10-secosteroids
US4920216A (en) 1987-05-28 1990-04-24 The Trustees Of Columbia In The City Of New York Selective chlorination of steroids and other substrates directed by covalently linked pyridine derivatives acting as templates
US4910192A (en) 1987-12-04 1990-03-20 Sri International Topically active steroidal anti-inflammatory agents
US4876250A (en) 1988-10-31 1989-10-24 Alcon Laboratories, Inc. Methods for controlling ocular hypertension with angiostatic steroids
US5990099A (en) 1988-10-31 1999-11-23 Alcon Laboratories, Inc. Angiostatic agents and methods and compositions for controlling ocular hypertension
WO1990012577A3 (en) 1989-04-18 1992-03-19 Upjohn Co Topical anti-angiogenic as hair growth inhibitors
WO1990015816A1 (en) 1989-06-16 1990-12-27 The Upjohn Company Suramin type compounds and angiostatic steroids to inhibit angiogenesis
US6090794A (en) 1990-04-19 2000-07-18 The General Hospital Corporation Inhibition of neurofibrosarcoma growth and angiogenesis
WO1991019731A1 (en) 1990-06-11 1991-12-26 The Upjohn Company Steroids which inhibit angiogenesis
US5972922A (en) 1990-06-11 1999-10-26 Alcon Laboratories, Inc. Steroids which inhibit angiogenesis
US5451690A (en) 1991-04-10 1995-09-19 Roussel Uclaf Lambda-1,4-pregnadiene-3,20-diones
US5434258A (en) 1991-04-10 1995-07-18 Roussel Uclaf δ1,4 -pregnadiene-3,20-diones
US5310896A (en) 1991-04-10 1994-05-10 Roussel Uclaf Process for preparing Δ1,4 -pregnadiene-3,20-diones
US5412091A (en) 1991-06-25 1995-05-02 Roussel Uclaf 16-methyl-Δ1,4-pregnadiene-3,20-dione
US5616743A (en) 1991-06-25 1997-04-01 Roussel Uclaf 16-methyl-Δ1,4 -pregnadiene-3,20-diones
US5248773A (en) 1991-06-25 1993-09-28 Roussel-Uclaf 16-methyl-Δ1,4-pregnadiene-3,20-diones
EP1236469A2 (en) 1991-11-22 2002-09-04 Alcon Laboratories, Inc. Angiostatic steroids
US5502183A (en) 1992-01-28 1996-03-26 Schering Corporation Steroid intermediates and processes for their preparation
WO1993025570A1 (en) 1992-06-09 1993-12-23 Richter Gedeon Vegyészeti Gyár Rt. Novel steroids with pregnane skeleton, pharmaceutical compositions containing them and process for preparing same
US6194565B1 (en) 1992-06-11 2001-02-27 Hoechst Marion Roussel Process for preparing 16 α-methyl-steroids
US5731447A (en) 1992-06-11 1998-03-24 Roussel Uclaf Process for preparing 16α-methyl-steroids
US20080234380A1 (en) 1992-06-30 2008-09-25 Shapiro Howard K Compositions and method for treatment of chronic inflammatory diseases
US20050090553A1 (en) 1992-06-30 2005-04-28 Shapiro Howard K. Compositions and method for treatment of chronic inflammatory diseases
US5508452A (en) 1993-01-20 1996-04-16 Roussel Uclaf Preparation of 16α-methyl steroids
WO1995018621A1 (en) 1994-01-04 1995-07-13 Duke University Methods of inhibiting angiogenesis and tumor growth, and treating ophthalmologic conditions with angiostatic and therapeutic steroids
US5502222A (en) 1994-06-01 1996-03-26 Schering Corporation Process for preparing delta 9,11 and 21-chloro corticosteroids
US5616742A (en) 1994-06-01 1997-04-01 Schering Corporation Process for preparing Δ9,11 and 21-chloro corticosteroids
US5750745A (en) 1994-06-01 1998-05-12 Schering Corporation Process for preparing delta 9, 11 and 21-chlorocorticosteroids
WO1996006618A1 (en) 1994-09-01 1996-03-07 Pharmacia & Upjohn Company Cosolvent parenteral formulation of tirilazad
WO1996010402A1 (en) 1994-10-04 1996-04-11 Knox Van Dyke Multi-faceted method to repress reproduction of latent viruses in humans and animals
WO1997022616A1 (en) 1995-12-20 1997-06-26 Schering Corporation PROCESS FOR PREPARATION OF 9,11β-EPOXIDE STEROIDS
US20020002155A1 (en) 1996-04-18 2002-01-03 Hoechst Marion Roussel Steroids, their use as medicaments, their preparation process, the intermediates of this process and the compositions containing them
WO1997039018A1 (en) 1996-04-18 1997-10-23 Hoechst Marion Roussel New pregnane derivatives with no alpha-17 substitutent, their medicinal use, manufacturing method and its intermediaries and related compounds
EP0875516A2 (en) 1997-04-30 1998-11-04 Farmabios S.r.l. Process for the preparation of 16,17 acetals of pregnane derivatives with control of the epimeric distribution at the C-22 position
US6169178B1 (en) 1997-04-30 2001-01-02 Farmabios S.R.L. Process for the preparation of 16,17 acetals of pregnane derivatives with control of the epimeric distribution at the C-22 position
WO1999003503A1 (en) 1997-07-16 1999-01-28 Alcon Laboratories, Inc. The use of angiostatic steroids in photodynamic therapy
US6011023A (en) 1997-08-27 2000-01-04 Alcon Laboratories, Inc. Angiostatic steroids
WO1999017778A1 (en) 1997-10-07 1999-04-15 Cephalon Inc. α-KETOAMIDE MULTICATALYTIC PROTEASE INHIBITORS
US6090798A (en) 1997-12-19 2000-07-18 Alcon Laboratories, Inc. Treatment of GLC1A glaucoma with glucocorticoid antagonists
WO1999032102A1 (en) 1997-12-19 1999-07-01 Alcon Laboratories, Inc. Treatment of glc1a glaucoma with glucocorticoid antagonists
US20030032630A1 (en) 1998-05-27 2003-02-13 Manuel Merlos Roca Use of a 2-hydroxy-4-trifluoromethylbenzoic acid derivatives as inhibitors of the activation of the nuclear transcription factor NF-kappaB
WO2000038653A1 (en) 1998-12-23 2000-07-06 Idea Ag. Improved formulation for topical non-invasive application in vivo
RU2156256C1 (en) 1999-12-20 2000-09-20 Андрюшина Валентина Александровна Method of preparing corticosteroids
WO2002043785A2 (en) 2000-11-29 2002-06-06 Oculex Pharmaceuticals, Inc. Intraocular implants for preventing transplant rejection in the eye
US20040181055A1 (en) 2001-07-26 2004-09-16 Ragactives, S.L. Stereoselective process for the production of 6alpha-fluorpregnanes and intermediates
WO2003014141A1 (en) 2001-08-10 2003-02-20 Cosmo S.P.A. 17alfa, 21-dihydroxypregnene esters as antiandrogenic agents
EP1336602A1 (en) 2002-02-13 2003-08-20 Giovanni Scaramuzzino Nitrate prodrugs able to release nitric oxide in a controlled and selective way and their use for prevention and treatment of inflammatory, ischemic and proliferative diseases
WO2004012700A2 (en) 2002-08-05 2004-02-12 Torrent Pharmaceuticals Limited Dosage form comprising high dose high soluble active ingredient as modified release and low dose active ingredient as immediate release
US20060018933A1 (en) 2002-08-05 2006-01-26 Navin Vaya Novel drug delivery system
US20060024365A1 (en) 2002-08-05 2006-02-02 Navin Vaya Novel dosage form
US20060153916A1 (en) 2002-08-05 2006-07-13 Navin Vaya Novel dosage form
WO2004012699A2 (en) 2002-08-05 2004-02-12 Torrent Pharmaceuticals Limited Modified release composition comprising coated micro matrix particles containing the high soluble active ingredient and a release controlling agent
CN1896090A (en) 2005-07-14 2007-01-17 天津药业研究院有限公司 Preparation of delta 9(11) steroid compound
CN1907999A (en) 2005-08-05 2007-02-07 天津天药药业股份有限公司 Preparation method for 9,11-de-esterification of metllylpredllisolone series products
US20070212751A1 (en) 2006-01-18 2007-09-13 Solvay Pharmaceuticals Gmbh Microbial method for the 11beta hydroxylation of 9beta, 10alpha-steriods
CN101347438A (en) 2007-07-20 2009-01-21 天津药业研究院有限公司 Medicament for slowing down angiogenesis of eye section
CN101353368A (en) 2007-07-27 2009-01-28 天津药业研究院有限公司 Pregna medicament for treating tumor
CN101397320A (en) 2007-09-29 2009-04-01 天津天药药业股份有限公司 Method for preparing dexamethasone and series products thereof
CN101397319A (en) 2007-09-29 2009-04-01 天津天药药业股份有限公司 Method for preparing betamethasone and series products thereof
CN101434631A (en) 2007-11-16 2009-05-20 天津金耀集团有限公司 Estra nitrate ester medicament for inhibiting angiogenesis
US8207151B2 (en) 2008-05-28 2012-06-26 Validus Biopharma Inc. Non-hormonal steroid modulators of NF-κB for treatment of disease
WO2009155056A2 (en) 2008-05-28 2009-12-23 Validus Genetics Non-hormonal steroid modulators of nf-kb for treatment of disease
WO2009155056A3 (en) 2008-05-28 2010-03-25 Validus Genetics Non-hormonal steroid modulators of nf-kb for treatment of disease
US20100087408A1 (en) 2008-05-28 2010-04-08 Validus Genetics NON-HORMONAL STEROID MODULATORS OF NF-kB FOR TREATMENT OF DISEASE
US8673887B2 (en) 2008-05-28 2014-03-18 Reveragen Biopharma, Inc Non-hormonal steroid modulators of NF-kB for treatment of disease
US8334279B2 (en) 2008-05-28 2012-12-18 Validus Genetics Non-hormonal steroid modulators of NF-κB for treatment of disease
CN101624414A (en) 2008-07-07 2010-01-13 天津金耀集团有限公司 Nitrate medicament for inhibiting angiogenesis
CN101759742A (en) 2008-11-06 2010-06-30 天津金耀集团有限公司 Preparation method of 16 Alpha-methyl steroidal compound
WO2011127048A3 (en) 2010-04-05 2012-02-02 Validus Biopharma NON-HORMONAL STEROID MODULATORS OF NF-ĸB FOR TREATMENT OF DISEASE
WO2011127048A2 (en) 2010-04-05 2011-10-13 Validus Biopharma NON-HORMONAL STEROID MODULATORS OF NF-ĸB FOR TREATMENT OF DISEASE

Non-Patent Citations (45)

* Cited by examiner, † Cited by third party
Title
Asahara et al., "High DNA-Binding Activity of Transcription Factor NF-Kappa B in Synovial Membranes Of Patients With Rheumatoid Arthritis", Biochemistry and Molecular Biology International (1995), 37(5), 827-32.
Australian Application No. 2009260471, Examination Report, dated Nov. 1, 2011.
Australian Application No. 2009260471, Response to First Examination Report, dated Oct. 30, 2013.
Australian Application No. 2009260471, Response to Second Examination Report, dated Nov. 20, 2014.
Australian Application No. 2009260471, Second Examination Report, dated Nov. 17, 2014.
Bao et al., "A Novel Antiinflammatory Role for Andrographolide in Asthma via Inhibition of the Nuclear Factor-kB Pathway", Am. J. Respir. Crit. Care Med., 2009, 179, 657-665.
Chinese Application No. 200980124859, Letter Reporting Second Office Action, dated Feb. 16, 2013.
Chinese Application No. 200980124859, Office Action dated Apr. 18, 2012.
De Meglio et al., "Amelioration of Acute Inflammation by Systemic Administration of a Cell-Permeable Peptide Inhibitor Of NF-Kb Activation", Arthritis & Rheumatism, 2005, 52(3), 951958.
FR-M335, Laboratoires Francias de Chimiotherapie, Oct. 1961, 4 pgs.
Hall et al., "Effects Of the 21-Amino Steroid U 74006F on Experimental Head Injury in Mice", Journal of Neurosurgery (1988), 68(3), 456-61.
Hori et al., "Differential Effects of Angiostatic Steroids and Dexamethasone on Angiogenesis and Cytokine Levels in Rat Sponge Implants", Br J. Pharmacol, 1996, 118,158491.
Jacobsen et al., "Novel 21-Aminosteroids That Inhibit Iron-Dependent Lipid Peroxidation and Protect Against Central Nervous System Trauma", Journal of Medicinal Chemistry (1990), 33(4), 1145-51.
Japanese Application No. 2011511822, Amended Claims and Remarks, dated Jun. 9, 2014.
Japanese Application No. 2011511822, English Translation of Official Action, dated Dec. 17, 2013.
Jia, Zhong et al., "Regulation of Oxygen Free Radical on Expression of Interleukin-8 in Gastric Cancer Cell Line SGC-7901", Zhongguo Zhongxiyi Jiehe Waike Zazhi, (2006), 12(3), 264-8.
Kalsi et al., "Suppressive Effects of a Novel Antioxidant Compound on Human T Cell Functions in Vitro", Agents Actions, 39(Special Conference Issue), 1993, C110-C112.
Korean Application No. 20107029408, English Translation of Notice of Grounds for Rejection, dated Mar. 31, 2014.
Korean Application No. 20107029408, Response to Office Action, dated Apr. 17, 2014.
Lingham et al. CAS: 130: 22070, 1998. *
Lu et al. CAS: 150: 222244, 2009. *
lzmailova et al, "Use of Molecular Imaging to Quantify Response to IKK-2 Inhibitor Treatment in Murine Arthritis", Arthritis & Rheumatism 2007, 56(1), 117-128.
McNatt et al, "Angiostatic Activity of Steroids in the Chick Embryo CAM and Rabbit Cornea Models of Neovascularization", Journal of Ocular Pharmacology and Therapeautics, 1999, 15(5), 413-423.
Metzinger et al., "Modulation by Prednisolone of Calcium Handling in Skeletal Muscle Cells", Br J. Pharmacol, 1995, 116, 2811-6.
Mexican Patent Application No. 2010012976, First Office Action Response, dated Jun. 4, 2013.
Mexican Patent Application No. 2010012976, First Office Action, dated Apr. 3, 2013.
Mexican Patent Application No. 2010012976, Letter Reporting First Office Action, dated Apr. 3, 2013.
Mexican Patent Application No. 2010012976, Response to Third Office Action, dated Aug. 6, 2014.
Mexican Patent Application No. 2010012976, Second Office Action, dated Oct. 2, 2013.
Mexican Patent Application No. 2010012976, Third Office Action, dated Mar. 11, 2013.
New Zealand Application no. 589444 - Response to Second Office Action.
New Zealand Application No. 589444, Examination Report, dated Dec. 20, 2011.
New Zealand Application No. 589444, First Official Action, dated Apr. 19, 2011.
New Zealand Application No. 589444, Notice of Acceptance, dated Mar. 5, 2013.
New Zealand Application No. 589444, Response to Office Action, dated Nov. 30, 2011.
New Zealand Application No. 589444, Second Official Action, dated Oct. 25, 2012.
New Zealand Application No. 603107, Notice of Acceptance, dated Mar. 20, 2014.
New Zealand Application No. 603107, Response to Office Action, dated Feb. 7, 2014.
New Zealand Application No. 63107, First Office Action, dated Oct. 25, 2012.
New Zealand Application No. 63107, Specification as filed, Oct. 18, 2012.
PCT Application No. PCT/US2009/045489, European Extended Search Report, dated Dec. 5, 2011.
Poynter et al., "NF-Kb Activation in Airways Modulates Allergic Inflammation But Not Hyperresponsiveness", J. Immunol., 2004, 173, 7003-7009.
Rousseau et al. CAS: 87: 161924, 1977. *
SciFinder Search Results, (May 28, 2009), 1-20.
Vernier et al., "Antioxidant Lazaroids Enhance Differentiation of C2 Skeletal Muscle Cells", Neuroscience Letters, 1995, 186, 177-80.

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10857161B2 (en) 2008-05-28 2020-12-08 Reveragen Biopharma, Inc. Non-hormonal steroid modulators of NF-kB for treatment of disease
US11833159B2 (en) 2008-05-28 2023-12-05 Reveragen Biopharma, Inc. Non-hormonal steroid modulators of NF-kB for treatment of disease
US20160375037A1 (en) * 2015-06-29 2016-12-29 Reveragen Biopharma, Inc. Non-hormonal steroid modulators of nf-kappa beta for treatment of disease
US10799514B2 (en) * 2015-06-29 2020-10-13 Reveragen Biopharma, Inc. Non-hormonal steroid modulators of NF-kappa beta for treatment of disease
US11690853B2 (en) 2015-06-29 2023-07-04 Reveragen Biopharma, Inc. Non-hormonal steroid modulators of NF-κβ for treatment of disease
US11382922B2 (en) 2019-03-07 2022-07-12 Reveragen Biopharma, Inc. Aqueous oral pharmaceutical suspension compositions
US11471471B2 (en) 2019-03-07 2022-10-18 Reveragen Biopharma, Inc. Aqueous oral pharmaceutical suspension compositions
WO2020191087A1 (en) 2019-03-18 2020-09-24 Ptc Therapeutics, Inc. Therapeutic combinations for use in treating a muscular dystrophy
EP4230196A1 (en) 2022-02-21 2023-08-23 Som Innovation Biotech, S.A. Compounds for use in the treatment of dystrophinopathies
WO2023156645A1 (en) 2022-02-21 2023-08-24 Som Innovation Biotech, S.A. Compounds for use in the treatment of dystrophinopathies

Also Published As

Publication number Publication date
EP2556083A2 (en) 2013-02-13
US20160060289A1 (en) 2016-03-03
US20130196962A1 (en) 2013-08-01
WO2011127048A3 (en) 2012-02-02
US10000525B2 (en) 2018-06-19
WO2011127048A2 (en) 2011-10-13
EP2556083A4 (en) 2013-12-04

Similar Documents

Publication Publication Date Title
US11833159B2 (en) Non-hormonal steroid modulators of NF-kB for treatment of disease
US10000525B2 (en) Non-hormonal steroid modulators of NF-κB for treatment of disease
US11690853B2 (en) Non-hormonal steroid modulators of NF-κβ for treatment of disease
US20220324902A1 (en) Non-hormonal steroid modulators of nf-kappa beta for treatment of disease

Legal Events

Date Code Title Description
AS Assignment

Owner name: REVERAGEN BIOPHARMA, INC., MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MCCALL, JOHN M;HOFFMAN, ERIC;NAGARAJU, KANNEBOYINA;SIGNING DATES FROM 20130327 TO 20130328;REEL/FRAME:030121/0009

AS Assignment

Owner name: FREERUN TECHNOLOGIES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FORBES, FRANK;FRANZ, BENJAMIN JARED;REEL/FRAME:030747/0745

Effective date: 20130426

AS Assignment

Owner name: REVERAGEN BIOPHARMA, INC, MARYLAND

Free format text: CHANGE OF NAME;ASSIGNOR:VALIDUS BIOPHARMA, INC.;REEL/FRAME:036132/0924

Effective date: 20110315

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2551); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 4

AS Assignment

Owner name: UNITED STATES GOVERNMENT, MARYLAND

Free format text: CONFIRMATORY LICENSE;ASSIGNOR:REVERAGEN BIOPHARMA, INC.;REEL/FRAME:063649/0541

Effective date: 20230413

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2552); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 8